US11464856B2 - Combination therapy using inhibitors of human growth and differentiation factor 15 (GDF-15) and immune checkpoint blockers - Google Patents

Combination therapy using inhibitors of human growth and differentiation factor 15 (GDF-15) and immune checkpoint blockers Download PDF

Info

Publication number
US11464856B2
US11464856B2 US15/765,176 US201615765176A US11464856B2 US 11464856 B2 US11464856 B2 US 11464856B2 US 201615765176 A US201615765176 A US 201615765176A US 11464856 B2 US11464856 B2 US 11464856B2
Authority
US
United States
Prior art keywords
hgdf
gdf
cells
antibody
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US15/765,176
Other languages
English (en)
Other versions
US20190160169A1 (en
Inventor
Jörg Wischhusen
Markus Haake
Reinhard Dummer
Matthias MEHLING
Tina SCHÄFER
Martina SELLE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Julius Maximilians Universitaet Wuerzburg
Original Assignee
Julius Maximilians Universitaet Wuerzburg
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=57121227&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US11464856(B2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from GBGB1517531.8A external-priority patent/GB201517531D0/en
Priority claimed from GBGB1607801.6A external-priority patent/GB201607801D0/en
Application filed by Julius Maximilians Universitaet Wuerzburg filed Critical Julius Maximilians Universitaet Wuerzburg
Assigned to JULIUS-MAXIMILIANS-UNIVERSITÄT WÜRZBURG reassignment JULIUS-MAXIMILIANS-UNIVERSITÄT WÜRZBURG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MEHLING, Matthias, SCHÄFER, Tina, SELLE, Martina, HAAKE, MARKUS, WISCHHUSEN, Jörg, DUMMER, Reinhard
Publication of US20190160169A1 publication Critical patent/US20190160169A1/en
Application granted granted Critical
Publication of US11464856B2 publication Critical patent/US11464856B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5064Endothelial cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells

Definitions

  • the present invention relates to uses of inhibitors of human Growth and Differentiation Factor 15 (GDF-15), and to combined uses of such inhibitors with immune checkpoint blockers, in the treatment of solid cancers.
  • GDF-15 human Growth and Differentiation Factor 15
  • growth factors including factors such as VEGF, PDGF, TGF- ⁇ and GDF-15.
  • GDF-15 growth and differentiation factor-15
  • TGF- ⁇ superfamily a protein which is intracellularly expressed as a precursor, subsequently processed and eventually becomes secreted from the cell into the environment.
  • Both the active, fully processed (mature) form and the precursor of GDF-15 can be found outside cells.
  • the precursor covalently binds via its COOH-terminal amino acid sequence to the extracellular matrix (Bauskin A R et al., Cancer Research 2005) and thus resides on the exterior of a cell.
  • the active, fully processed (mature) form of GDF-15 is soluble and is found in blood sera.
  • the processed form of GDF-15 may potentially act on any target cell within the body that is connected to the blood circulation, provided that the potential target cell expresses a receptor for the soluble GDF-15 ligand.
  • GDF-15 is found under physiological conditions in the placenta.
  • malignant cancers especially aggressive brain cancers, melanoma, lung cancer, gastrointestinal tumors, colon cancer, pancreatic cancer, prostate cancer and breast cancer (Mimeault M and Batra S K, J. Cell Physiol 2010)
  • GDF-15 levels exhibit increased GDF-15 levels in the tumor as well as in blood serum.
  • correlations have been described between high GDF-15 expression and chemoresistance (Huang C Y et al., Clin. Cancer Res. 2009) and between high GDF-15 expression and poor prognosis, respectively (Brown D A et al., Clin. Cancer Res. 2009).
  • GDF-15 is expressed in gliomas of different WHO grades as assessed by immunohistochemistry (Roth et al., Clin. Cancer Res. 2010). Further, Roth et al. stably expressed short hairpin RNA-expressing DNA constructs targeting endogenous GDF-15 or control constructs in SMA560 glioma cells. When using these pre-established stable cell lines, they observed that tumor formation in mice bearing GDF-15 knockdown SMA560 cells was delayed compared to mice bearing control constructs.
  • Patent applications WO 2005/099746 and WO 2009/021293 relate to an anti-human-GDF-15 antibody (Mab26) capable of antagonizing effects of human GDF-15 (hGDF-15) on tumor-induced weight loss in vivo in mice.
  • Mc26 an anti-human-GDF-15 antibody
  • hGDF-15 human GDF-15
  • Johnen H et al. reported effects of an anti-human-GDF-15 monoclonal antibody on cancer-induced anorexia and weight loss but did not observe any effects of the anti-human-GDF-15 antibody on the size of the tumor formed by the cancer.
  • WO 2014/049087 and PCT/EP2015/056654 relate to monoclonal antibodies to hGDF-15 and medical uses thereof.
  • immune checkpoint blockers such as inhibitors of human PD-1 and inhibitors of human PD-L1.
  • a rationale behind the use of these immune checkpoint blockers is that by blocking immune checkpoints which prevent the immune system from targeting cancer antigens and the respective cancer cells, an immune response to the cancer may be more effective. While immune checkpoint blockers as well as particular combinations of immune checkpoint blockers have been shown improve patient survival in melanoma patients (Cully M, “Combinations with checkpoint inhibitors at wavefront of cancer immunotherapy.”, Nat Rev Drug Discov. 2015 June; 14(6):374-5.), not all melanoma patients exhibited a complete response, and results for many other cancers are yet to be disclosed, still there are reasons (like the mutational burden) which suggest that results in other indications will be less favorable.
  • an inhibitor of hGDF-15 can be used to inhibit the negative effects of hGDF-15 on the patients' responses to the treatment with immune checkpoint blockers, and to improve the patients' responses to the treatment with immune checkpoint blockers.
  • hGDF-15 therapeutic inhibition of hGDF-15 can be used to increase the percentage of CD8 + T lymphocytes in solid cancers including tumor metastases.
  • This increase of CD8 + T lymphocytes in the solid cancers can favorably be used for therapy, in particular immunotherapy, of the solid cancers.
  • a particularly favorable therapeutic combination is the combination of an hGDF-15 inhibitor with an immune checkpoint blocker.
  • An advantageous effect of this combination is that inhibition of hGDF-15 will increase the percentage of CD8 + T lymphocytes in the solid cancers and thereby lead to a synergistic therapeutic effect with immune checkpoint inhibition.
  • hGDF-15 inhibitors can increase the percentage of CD8 + T lymphocytes in the solid cancers. Therefore, according to the invention, a treatment with hGDF-15 inhibitors can be used to increase adhesion of T cells including CD8 + T cells to endothelial cells. Such treatment according to the invention will increase entry of CD8 + T cells from the blood stream into solid cancers.
  • the increased percentage of CD8 + T cells in solid cancers which will result from such treatment with hGDF-15 inhibitors, is advantageous for, and can be used in, cancer therapy, e.g. cancer immunotherapy.
  • hGDF-15 inhibitors Since the entry of CD8 + T cells into solid cancers and the presence of these CD8 + T cells in the solid cancers is particularly advantageous for therapeutic approaches using immune checkpoint blockers, a particularly advantageous use of hGDF-15 inhibitors according to the invention is their use in combination with immune checkpoint blockers.
  • the present invention provides improved means for cancer therapy by providing the preferred embodiments described below:
  • FIG. 1 This Figure shows the GDF-15 serum levels for responders and non-responders to the treatment regimen.
  • FIG. 2 This Figure shows the numbers of responders and non-responders in the patient groups having hGDF-15 serum levels of ⁇ 1.8 ng/ml, 1.8-4.2 ng/ml, and >4.2 ng/ml, respectively.
  • FIG. 3 Probability of response to treatment (responder 1) as predicted by the Generalized Linear Model using GDF-15 as continuous predictor. Circles show the data, the curve shows the model. The vertical line indicates the GDF-15 concentration where the probability of treatment response is 0.5.
  • FIG. 4 Kaplan-Meier curves for survival in the three groups defined by the GDF-15 serum level ( ⁇ 1.8, 1.8-4.2, >4.2 ng/ml).
  • FIGS. 5A-5B FIG. 5A : Probability of response to treatment (responder 1) as predicted by the Generalized Linear Model model using LDH as continuous predictor. Circles show the data, the curve shows the model. The vertical line indicates the LDF concentration where the probability of treatment response is 0.5. The patient cohort was identical. However, reliable determination of LDH levels failed in four patients due to hemolysis.
  • FIG. 5B Graphical representation of responders and non-responders and their respective hGDF-15 and LDH levels. When cut-off values are selected to cover all responders, testing based on GDF-15 allows for identification of 6 (out of 9) non-responders whereas analyses based on LDH levels can only discriminate 4 (out of 9) non-responders. For LDH testing, 4 hemolytic samples had to be excluded which causes loss of data.
  • FIG. 6 This Figure shows exemplary tissue sections from melanoma brain metastases having no (upper panel) or high (lower panel) GDF-15 immunoreactivity, which were stained by immunohistochemistry for GDF-15 and for the T-cell marker proteins CD3 and CD8, respectively, as indicated in the Figure.
  • CD3 and CD8-positive cells are indicated by arrows in the high GDF-15 samples.
  • the CD3 and CD8 stainings were made from the same area of serial sections (however not from the identical section).
  • FIGS. 7A-7B This Figure shows a plot of the percentage of CD3 + cells against the GDF-15 score across different melanoma brain metastases ( 7 A) and a plot of the percentage of CD8 + cells against the GDF-15 score across different melanoma brain metastases ( 7 B).
  • FIG. 8 This Figure shows a plot of the GDF-15 score against the percentage of CD8 + and CD3 + T cells, respectively, in brain metastases from different tumor entities (melanoma, CRC, RCC, NSCLC and SCLC).
  • FIGS. 9A-9D show the number of rolling T cells per field of view per second. Data were obtained from channel #3 (“GDF-15”) and channel #2 (“control”).
  • FIG. 9B shows the rolling speed of the T cells (measured in pixels per 0.2 seconds). Data were obtained from channel #3 (“GDF-15”) and channel #2 (“control”).
  • FIG. 9C shows the number of adhering cells per field of view. Data were obtained from channel #3 (“GDF-15”) and channel #2 (“control”).
  • FIG. 9D shows the number of adhering cells per field of view. Data were obtained from channel #3 (“GDF-15”) and channel #2 (“control”).
  • FIGS. 10A-10B show the number of rolling T cells per field of view per second. Data were obtained from channel #1 (control T cells on unstimulated HUVEC as “neg. control”), channel #2 (control T cells on stimulated HUVEC as “pos. control”), channel #3 (“GDF-15”) channel #4 (“UACC 257”: T cells cultured in the supernatant of UACC 257 melanoma cells containing secreted GDF-15) and channel #5 (“UACC257+
  • FIG. 10B The flow/adhesion assay was conducted as described in Example 3. T-cells were pre-incubated with 100 ng/ml GDF-15 for 1 hour or with 100 ng/ml GDF-15, which was pre-incubated with 10 ⁇ g/ml antibody for 1 hour, as indicated.
  • Anti-GDF-15 antibodies were used: H1L5 (Humanized B1-23), 01G06 and 03G05 (Humanized Anti-GDF-15 Antibodies Engineered According to Sequences from WO 2014/100689). The results are shown in the Figure, which shows the number of rolling cells per field of view per 20 seconds.
  • FIG. 11 C57BL/6J mice were subcutaneously inoculated with 2 ⁇ 10 5 colon MC38 tghGDF-15 cells. Treatment with anti GDF-15 antibody (20 mg/kg of body weight) was initiated on day 0 and repeated on days 3, 7, 10, 14, 17, and 21. On day 13, animals bearing similarly sized tumors (100-150 mm 3 ) were either treated or not with Poly-ICLC (also abbreviated as “Poly-IC”) and anti CD40 antibody. Mice rejecting the pre-established tumor were followed for 57 days. Tumor-bearing mice were sacrificed according to the criteria for animal welfare.
  • FIG. 12 Cumulative survival in patient groups having GDF-15 levels of ⁇ 1.5 ng/ml and ⁇ 1.5 ng/ml, respectively.
  • FIG. 13 Cumulative survival in patient groups having high GDF-15 levels (i.e. the 50 patients with the highest GDF-15 levels) and low GDF-15 levels (i.e. the 49 patients with the lowest GDF-15 levels), respectively (median split of the total study cohort).
  • FIGS. 14A-14B hGDF-15 Serum Levels do not Significantly Correlate with the Mutational Burden of the Tumors.
  • FIG. 14A shows a plot for cancer patient data obtained from the Cancer Genome Atlas (TGCA) considering only patients with high-grade malignant melanoma (the Cancer Genome Atlas is described in the reference of Cheng P F et al.: Data mining The Cancer Genome Atlas in the era of precision cancer medicine. Swiss Med Wkly.
  • FIG. 14B shows a plot for cancer patient data from 40 additional metastatic malignant melanoma patients from the University Hospital Zurich, which were separately analyzed.
  • FIG. 15 Immunocytochemistry pictures for CD8a in mice harboring wild-type tumors or tumors overexpressing transgenic (tg) hGDF15 are shown. Tissue sections were stained with anti-CD8a (1:100 dilution; 4SM15 antibody purchased from eBioscience).
  • antibody refers to any functional antibody that is capable of specific binding to the antigen of interest, as generally outlined in chapter 7 of Paul, W. E. (Ed.).: Fundamental Immunology 2nd Ed. Raven Press, Ltd., New York 1989, which is incorporated herein by reference.
  • the term “antibody” encompasses antibodies from any appropriate source species, including chicken and mammalian such as mouse, goat, non-human primate and human.
  • the antibody is a humanized antibody.
  • the antibody is preferably a monoclonal antibody which can be prepared by methods well-known in the art.
  • antibody encompasses an IgG-1, -2, -3, or -4, IgE, IgA, IgM, or IgD isotype antibody.
  • antibody encompasses monomeric antibodies (such as IgD, IgE, IgG) or oligomeric antibodies (such as IgA or IgM).
  • antibody also encompasses—without particular limitations—isolated antibodies and modified antibodies such as genetically engineered antibodies, e.g. chimeric antibodies.
  • each monomer of an antibody comprises two heavy chains and two light chains, as generally known in the art.
  • each heavy and light chain comprises a variable domain (termed V H for the heavy chain and V L for the light chain) which is important for antigen binding.
  • V H variable domain
  • V L variable domain
  • These heavy and light chain variable domains comprise (in an N-terminal to C-terminal order) the regions FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4 (FR, framework region; CDR, complementarity determining region which is also known as hypervariable region).
  • a “monoclonal antibody” is an antibody from an essentially homogenous population of antibodies, wherein the antibodies are substantially identical in sequence (i.e. identical except for minor fraction of antibodies containing naturally occurring sequence modifications such as amino acid modifications at their N- and C-termini). Unlike polyclonal antibodies which contain a mixture of different antibodies directed to numerous epitopes, monoclonal antibodies are directed to the same epitope and are therefore highly specific.
  • the term “monoclonal antibody” includes (but is not limited to) antibodies which are obtained from a monoclonal cell population derived from a single cell clone, as for instance the antibodies generated by the hybridoma method described in Kohler and Milstein (Nature, 1975 Aug.
  • a monoclonal antibody may also be obtained from other suitable methods, including phage display techniques such as those described in Clackson et al. (Nature. 1991 Aug. 15; 352(6336):624-8) or Marks et al. (J Mol Biol. 1991 Dec. 5; 222(3):581-97).
  • a monoclonal antibody may be an antibody that has been optimized for antigen-binding properties such as decreased Kd values, optimized association and dissociation kinetics by methods known in the art.
  • Kd values may be optimized by display methods including phage display, resulting in affinity-matured monoclonal antibodies.
  • the term “monoclonal antibody” is not limited to antibody sequences from particular species of origin or from one single species of origin. Thus, the meaning of the term “monoclonal antibody” encompasses chimeric monoclonal antibodies such as humanized monoclonal antibodies.
  • Humanized antibodies are antibodies which contain human sequences and a minor portion of non-human sequences which confer binding specificity to an antigen of interest (e.g. human GDF-15).
  • humanized antibodies are generated by replacing hypervariable region sequences from a human acceptor antibody by hypervariable region sequences from a non-human donor antibody (e.g. a mouse, rabbit, rat donor antibody) that binds to an antigen of interest (e.g. human GDF-15).
  • framework region sequences of the acceptor antibody may also be replaced by the corresponding sequences of the donor antibody.
  • a “humanized antibody” may either contain other (additional or substitute) residues or sequences or not.
  • Such other residues or sequences may serve to further improve antibody properties such as binding properties (e.g. to decrease Kd values) and/or immunogenic properties (e.g. to decrease antigenicity in humans).
  • binding properties e.g. to decrease Kd values
  • immunogenic properties e.g. to decrease antigenicity in humans.
  • Non-limiting examples for methods to generate humanized antibodies are known in the art, e.g. from Riechmann et al. (Nature. 1988 Mar. 24; 332(6162):323-7) or Jones et al. (Nature. 1986 May 29-Jun. 4; 321(6069):522-5).
  • human antibody relates to an antibody containing human variable and constant domain sequences. This definition encompasses antibodies having human sequences bearing single amino acid substitutions or modifications which may serve to further improve antibody properties such as binding properties (e.g. to decrease Kd values) and/or immunogenic properties (e.g. to decrease antigenicity in humans).
  • human antibody excludes humanized antibodies where a portion of non-human sequences confers binding specificity to an antigen of interest.
  • an “antigen-binding portion” of an antibody as used herein refers to a portion of an antibody that retains the capability of the antibody to specifically bind to the antigen (e.g. hGDF-15, PD-1 or PD-L1). This capability can, for instance, be determined by determining the capability of the antigen-binding portion to compete with the antibody for specific binding to the antigen by methods known in the art.
  • the antigen-binding portion may contain one or more fragments of the antibody.
  • the antigen-binding portion can be produced by any suitable method known in the art, including recombinant DNA methods and preparation by chemical or enzymatic fragmentation of antibodies.
  • Antigen-binding portions may be Fab fragments, F(ab′) fragments, F(ab′) 2 fragments, single chain antibodies (scFv), single-domain antibodies, diabodies or any other portion(s) of the antibody that retain the capability of the antibody to specifically bind to the antigen.
  • an “antibody” e.g. a monoclonal antibody
  • an “antigen-binding portion” may have been derivatized or be linked to a different molecule.
  • molecules that may be linked to the antibody are other proteins (e.g. other antibodies), a molecular label (e.g. a fluorescent, luminescent, colored or radioactive molecule), a pharmaceutical and/or a toxic agent.
  • the antibody or antigen-binding portion may be linked directly (e.g. in form of a fusion between two proteins), or via a linker molecule (e.g. any suitable type of chemical linker known in the art).
  • the terms “binding” or “bind” refer to specific binding to the antigen of interest (e.g. human GDF-15).
  • the Kd value is less than 100 nM, more preferably less than 50 nM, still more preferably less than 10 nM, still more preferably less than 5 nM and most preferably less than 2 nM.
  • an antibody or antigen-binding portion thereof which is “capable to compete” with a second antibody capable of binding to human GDF-15 means that said (first) antibody or antigen-binding portion thereof which is “capable to compete” is capable to reduce the binding of a 10 nM reference solution of the second antibody to human or recombinant human GDF-15 by 50%.
  • “capable to compete” means that the concentration of the (first) antibody or antigen-binding portion thereof that is needed in order to reduce the binding of the 10 nM reference solution of the second antibody to human or recombinant human GDF-15 by 50% is less than 1000 nM, preferably less than 100 nM and more preferably less than 10 nM.
  • the binding is measured by surface plasmon resonance measurements or by Enzyme-linked Immunosorbent assay (ELISA) measurements, preferably by surface plasmon resonance measurements.
  • epitope refers to a small portion of an antigen that forms the binding site for an antibody.
  • binding or competitive binding of antibodies or their antigen-binding portions to the antigen of interest is preferably measured by using surface plasmon resonance measurements as a reference standard assay, as described below.
  • K D or “K D value” relate to the equilibrium dissociation constant as known in the art. In the context of the present invention, these terms relate to the equilibrium dissociation constant of an antibody with respect to a particular antigen of interest (e.g. human GDF-15)
  • the equilibrium dissociation constant is a measure of the propensity of a complex (e.g. an antigen-antibody complex) to reversibly dissociate into its components (e.g. the antigen and the antibody).
  • K D values are preferably determined by using surface plasmon resonance measurements as described below.
  • an “isolated antibody” as used herein is an antibody that has been identified and separated from the majority of components (by weight) of its source environment, e.g. from the components of a hybridoma cell culture or a different cell culture that was used for its production (e.g. producer cells such as CHO cells that recombinantly express the antibody). The separation is performed such that it sufficiently removes components that may otherwise interfere with the suitability of the antibody for the desired applications (e.g. with a therapeutic use of the anti-human GDF-15 antibody according to the invention).
  • Methods for preparing isolated antibodies are known in the art and include Protein A chromatography, anion exchange chromatography, cation exchange chromatography, virus retentive filtration and ultrafiltration.
  • the isolated antibody preparation is at least 70% pure (w/w), more preferably at least 80% pure (w/w), still more preferably at least 90% pure (w/w), still more preferably at least 95% pure (w/w), and most preferably at least 99% pure (w/w), as measured by using the Lowry protein assay.
  • a “diabody” as used herein is a small bivalent antigen-binding antibody portion which comprises a heavy chain variable domain linked to a light chain variable domain on the same polypeptide chain linked by a peptide linker that is too short to allow pairing between the two domains on the same chain. This results in pairing with the complementary domains of another chain and in the assembly of a dimeric molecule with two antigen binding sites.
  • Diabodies may be bivalent and monospecific (such as diabodies with two antigen binding sites for human GDF-15), or may be bivalent and bispecific (e.g. diabodies with two antigen binding sites, one being a binding site for human GDF-15, and the other one being a binding site for a different antigen). A detailed description of diabodies can be found in Holliger P et al. (““Diabodies”: small bivalent and bispecific antibody fragments.” Proc Natl Acad Sci USA. 1993 Jul. 15; 90(14):6444-8).
  • a “single-domain antibody” (which is also referred to as “NanobodyTM”) as used herein is an antibody fragment consisting of a single monomeric variable antibody domain. Structures of and methods for producing single-domain antibodies are known from the art, e.g. from Holt L J et al. (“Domain antibodies: proteins for therapy.” Trends Biotechnol. 2003 November; 21(11):484-90), Saerens D et al. (“Single-domain antibodies as building blocks for novel therapeutics.” Curr Opin Pharmacol. 2008 October; 8(5):600-8. Epub 2008 Aug. 22), and Arbabi Ghahroudi M et al. (“Selection and identification of single domain antibody fragments from camel heavy-chain antibodies.” FEBS Lett. 1997 Sep. 15; 414(3):521-6).
  • cancer and “cancer cell” is used herein in accordance with their common meaning in the art (see for instance Weinberg R. et al.: The Biology of Cancer. Garland Science: New York 2006. 850p).
  • solid cancers are solid cancers.
  • a “solid cancer” is a cancer which forms one or more solid tumors. Such solid cancers forming solid tumors are generally known in the art.
  • the term “solid cancer” encompasses both a primary tumor formed by the cancer and possible secondary tumors, which are also known as metastases.
  • Preferred solid cancers to be treated according to the invention are selected from the group consisting of melanoma, colorectal cancer, prostate cancer, head and neck cancer, urothelial cancer, stomach cancer, pancreatic cancer, liver cancer, testis cancer, ovarian cancer, endometrial cancer, cervical cancer, brain cancer, breast cancer, gastric cancer, renal cell carcinoma, Ewing's sarcoma, non-small cell lung cancer and small cell lung cancer, preferably selected from the group consisting of melanoma, colorectal cancer, prostate cancer, head and neck cancer, urothelial cancer, stomach cancer, pancreatic cancer, liver cancer, testis cancer, ovarian cancer, endometrial cancer and cervical cancer, more preferably selected from the group consisting of melanoma, colorectal cancer, prostate cancer, head and neck cancer, urothelial cancer and stomach cancer, and most preferably selected from the group consisting of melanoma, colorectal cancer and prostate cancer.
  • brain cancer refers to all brain cancers known in the art. It includes but is not limited to glioma (WHO grade I to IV), astrocytoma, meningioma and medulloblastoma.
  • head and neck cancer refers to all head and neck cancers known in the art. It includes but is not limited to oesophagus carcinoma, oral squamous cell carcinoma and hypopharyngeal cancer.
  • a particularly preferred head and neck cancer to be treated according to the invention is oral squamous cell carcinoma.
  • cancer growth as used herein relates to any measureable growth of the cancer.
  • cancer growth relates to a measurable increase in tumor volume over time. If the cancer has formed only a single tumor, “cancer growth” relates only to the increase in volume of the single tumor. If the cancer has formed multiple tumors such as metastases, “cancer growth” relates to the increase in volume of all measurable tumors.
  • the tumor volume can be measured by any method known in the art, including magnetic resonance imaging and computed tomography (CT scan).
  • Terms such as “treatment of cancer” or “treating cancer” according to the present invention refer to a therapeutic treatment.
  • An assessment of whether or not a therapeutic treatment works can, for instance, be made by assessing whether the treatment inhibits cancer growth in the treated patient or patients.
  • the inhibition is statistically significant as assessed by appropriate statistical tests which are known in the art.
  • Inhibition of cancer growth may be assessed by comparing cancer growth in a group of patients treated in accordance with the present invention to a control group of untreated patients, or by comparing a group of patients that receive a standard cancer treatment of the art plus a treatment according to the invention with a control group of patients that only receive a standard cancer treatment of the art.
  • treating cancer includes an inhibition of cancer growth where the cancer growth is inhibited partially (i.e. where the cancer growth in the patient is delayed compared to the control group of patients), an inhibition where the cancer growth is inhibited completely (i.e. where the cancer growth in the patient is stopped), and an inhibition where cancer growth is reversed (i.e. the cancer shrinks).
  • an assessment of whether or not a therapeutic treatment works can be made based on a classification of responders and non-responders by using the response evaluation criteria in solid tumours, version 1.1 (RECIST v1.1) (Eisenhauer et al.: New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). In: Eur. J. Cancer. 45, No. 2, January 2009, pp. 228-47).
  • an assessment of whether or not a therapeutic treatment works can be made based on known clinical indicators of cancer progression.
  • the treatment of cancer according to the invention can be a first-line therapy, a second-line therapy or a third-line therapy or a therapy that is beyond third-line therapy.
  • the meaning of these terms is known in the art and in accordance with the terminology that is commonly used by the US National Cancer Institute.
  • a treatment of cancer according to the present invention does not exclude that additional or secondary therapeutic benefits also occur in patients.
  • an additional or secondary benefit may be an influence on cancer-induced weight loss.
  • the primary treatment for which protection is sought is for treating the cancer itself, any secondary or additional effects only reflect optional, additional advantages of the treatment of cancer growth.
  • cancer immunotherapy is known in the art and generally relates to a treatment of cancer in which the immune system of the patient is used to treat the cancer.
  • Cancer cells harbor genomic mutations which give rise to cancer cell antigens that are specific to the cancer cells and different from the antigens of non-cancerous cells.
  • a cancer immunotherapy is a cancer immunotherapy wherein such cancer cell antigens are recognized by the immune system, and wherein cancer cells expressing these antigens are killed by the immune system.
  • cancer cells expressing these cancer cell antigens can be killed by CD8 + T-cells of the immune system.
  • a cancer immunotherapy can be assessed by immunomonitoring methods known in the art, e.g. by measuring intracellular IFN- ⁇ expression (e.g. in CD8 + T-cells and/or NK cells) in blood samples, measuring CD107a cell surface expression (e.g. on CD8 + T-cells and/or NK cells) in blood samples, measuring intracellular TNF- ⁇ expression (e.g. on leukocytes) in blood samples, intracellular Interleukin-2 expression (e.g. in CD8 + T-cells and/or in CD4 + T-cells) in blood samples, CD154 cell surface expression (e.g.
  • a “cancer immunotherapy” optionally encompasses a treatment where in addition to the immune system which is used to treat the cancer, additional mechanisms of cancer treatment are used.
  • additional mechanisms of cancer treatment are used.
  • a cancer immunotherapy by hGDF-15 inhibitors in human patients can also encompass additional treatment effects of hGDF-15 inhibitors which are independent from the immune system.
  • Another example of a cancer immunotherapy where additional mechanisms of cancer treatment can be used is a combination therapy with known chemotherapeutic agent(s).
  • Such combination therapy with known chemotherapeutic agent(s) may, for instance, not only include the treatment of cancer in which the immune system is used to treat the cancer but also include a treatment of cancer in which the cancer cells are killed by said chemotherapeutic agent(s) directly.
  • the term “increasing the percentage of CD8 + T-cells in a solid cancer” relates to any measurable increase in the percentage of CD8 + T-cells (i.e. the percentage of CD8 + T-cells calculated with respect to all cells) in the tumor or tumors formed by the solid cancer.
  • the increase is statistically significant as assessed by appropriate statistical tests which are known in the art.
  • An increase in the percentage of CD8 + T-cells in the tumor or tumors formed by the solid cancer can be determined by known methods for analyses of CD8 + T-cells in solid tumors. Such methods include analyses of tumor biopsies for CD8 + T-cells, e.g.
  • analyses of such tumor biopsies by immunohistochemistry using antibodies against CD8 and using a staining for the total number of cells may be assessed by comparing the percentages of CD8 + T-cells in tumors of a group of patients treated in accordance with the present invention to a control group of untreated patients, or by comparing a group of patients that receive a standard cancer treatment of the art plus a treatment according to the invention with a control group of patients that only receive a standard cancer treatment of the art.
  • CD8 + T-cells are preferably cells which endogenously occur in the human patient.
  • hGDF-15 serum levels can be measured by any methods known in the art.
  • a preferred method of measuring hGDF-15 serum levels is a measurement of hGDF-15 serum levels by Enzyme-Linked Immunosorbent Assay (ELISA) by using antibodies to GDF-15.
  • ELISA Enzyme-Linked Immunosorbent Assay
  • hGDF-15 serum levels may be determined by known electrochemiluminesence immunoassays using antibodies to GDF-15.
  • the Roche Elecsys® technology can be used for such electrochemiluminesence immunoassays.
  • the patient to be treated according to the invention is preferably a patient with elevated hGDF-15 serum levels.
  • elevated hGDF-15 serum levels means that the human patient has higher hGDF-15 levels in blood serum prior to administration of the hGDF-15 inhibitor according to the invention, when compared to median hGDF-15 levels in blood sera of healthy human control individuals as a reference.
  • the median hGDF-15 serum level of healthy human control individuals is ⁇ 0.8 ng/ml.
  • the expected range is between 0.2 ng/ml and 1.2 ng/ml in healthy human controls (Reference: Tanno T et al.: “Growth differentiation factor 15 in erythroid health and disease.” Curr Opin Hematol. 2010 May; 17(3): 184-190).
  • a patient to be treated according to the invention is a patient who has a hGDF-15 serum level of at least 1.2 ng/ml prior to the start of administration of the hGDF-15 inhibitor, preferably a patient who has a hGDF-15 serum level of at least 1.5 ng/ml prior to the start of administration of the hGDF-15 inhibitor, and more preferably a patient who has a hGDF-15 serum level of at least 1.8 ng/ml prior to the start of administration of the hGDF-15 inhibitor.
  • a patient to be treated according to the invention is a patient who has a hGDF-15 serum level of at least 1.2 ng/ml and not more than 12 ng/ml prior to the start of administration of the hGDF-15 inhibitor, preferably a patient who has a hGDF-15 serum level of at least 1.5 ng/ml and not more than 12 ng/ml prior to the start of administration of the hGDF-15 inhibitor, and more preferably a patient who has a hGDF-15 serum level of at least 1.8 ng/ml and not more than 12 ng/ml prior to the start of administration of the hGDF-15 inhibitor.
  • a patient to be treated according to the invention is a patient who has a hGDF-15 serum level of at least 1.2 ng/ml and not more than 10 ng/ml prior to the start of administration of the hGDF-15 inhibitor, preferably a patient who has a hGDF-15 serum level of at least 1.5 ng/ml and not more than 10 ng/ml prior to the start of administration of the hGDF-15 inhibitor, and more preferably a patient who has a hGDF-15 serum level of at least 1.8 ng/ml and not more than 10 ng/ml prior to the start of administration of the hGDF-15 inhibitor.
  • a patient to be treated according to the invention is a patient who has a hGDF-15 serum level of at least 1.2 ng/ml and not more than 8 ng/ml prior to the start of administration of the hGDF-15 inhibitor, preferably a patient who has a hGDF-15 serum level of at least 1.5 ng/ml and not more than 8 ng/ml prior to the start of administration of the hGDF-15 inhibitor, and more preferably a patient who has a hGDF-15 serum level of at least 1.8 ng/ml and not more than 8 ng/ml prior to the start of administration of the hGDF-15 inhibitor.
  • a patient to be treated according to the invention is a patient who has a hGDF-15 serum level of at least 2 ng/ml, at least 2.2 ng/ml, at least 2.4 ng/ml, at least 2.6 ng/ml, at least 2.8 ng/ml, at least 3.0 ng/ml, at least 3.2 ng/ml, at least 3.4 ng/ml, at least 3.6 ng/ml, at least 3.8 ng/ml, at least 4.0 ng/ml, or at least 4.2 ng/ml prior to the start of administration of the hGDF-15 inhibitor.
  • the patient is preferably a patient who has a hGDF-15 serum level of not more than 12 ng/ml prior to the start of administration of the hGDF-15 inhibitor. More preferably, in this embodiment, the patient is a patient who has a hGDF-15 serum level of not more than 10 ng/ml prior to the start of administration of the hGDF-15 inhibitor. Most preferably, in this embodiment, the patient is a patient who has a hGDF-15 serum level of not more than 8 ng/ml prior to the start of administration of the hGDF-15 inhibitor.
  • prior to the start of administration means the period of time immediately before administration of the hGDF-15 inhibitor according to the invention.
  • the term “prior to the start of administration” means a period of 30 days immediately before administration; most preferably a period of one week immediately before administration.
  • the hGDF-15 inhibitors and the immune checkpoint blockers used according to the invention can be administered by using methods known in the art. Such methods will be selected by the skilled person based on well-known considerations, including the chemical nature of the respective inhibitor (e.g. depending on whether the inhibitor is a short interfering RNA or an antibody).
  • Administration of known immune checkpoint blockers may be based on known administration schemes of these immune checkpoint blockers. For instance, administration of the immune checkpoint blockers may be based on the administration schemes used in the KEYNOTE-006 trial (C. Robert et al. N Engl J Med 2015; 372:2521-2532).
  • each occurrence of the term “comprising” may optionally be substituted with the term “consisting of”.
  • an “hGDF-15 inhibitor” can be any molecule which is capable of specifically inhibiting the function of human GDF-15 (hGDF-15).
  • hGDF-15 inhibitor is a molecule which specifically downregulates the expression of hGDF-15 and thereby inhibits hGDF-15 function.
  • a short interfering RNA or an siRNA hairpin construct can be used to specifically downregulate the expression of hGDF-15 and to inhibit hGDF-15 function.
  • Rules for the design and selection of short interfering RNA and siRNA hairpin construct sequences are known in the art and have for example been reviewed in Jackson and Linsley, Recognizing and avoiding siRNA off-target effects for target identification and therapeutic application, Nat Rev Drug Discov. 2010 January; 9(1):57-67.
  • Short interfering RNAs and siRNA hairpin constructs can be delivered to the human patients by any suitable methods, including viral delivery methods (as, for instance, reviewed in Knoepfel S A et al., “Selection of RNAi-based inhibitors for anti-HIV gene therapy.” World J Virol. 2012 Jun. 12; 1(3):79-90) and other delivery methods such as methods using conjugate groups which facilitate delivery into the cells (as, for instance, reviewed in Kanasty R et al., “Delivery materials for siRNA therapeutics.”, Nat Mater. 2013 November; 12(11):967-77.)
  • Whether or not a substance of interest is a “hGDF-15 inhibitor” can be determined by using the methods disclosed herein, as detailed in the preferred embodiments.
  • a preferred method in accordance with the preferred embodiments is the method used in Example 3.
  • the hGDF-15 inhibitor to be used is an antibody capable of binding to hGDF-15, or an antigen-binding portion thereof.
  • the antibody is a monoclonal antibody capable of binding to hGDF-15, or an antigen-binding portion thereof.
  • the hGDF-15 inhibitor in accordance with the invention is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 5 or an amino acid sequence at least 90% identical thereto, and wherein the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7 or an amino acid sequence at least 85% identical thereto.
  • the antibody or antigen-binding portion thereof comprises a heavy chain variable domain which comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 3 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 4, and the antibody or antigen-binding portion thereof comprises a light chain variable domain which comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 6, and a CDR2 region comprising the amino acid sequence ser-ala-ser.
  • the hGDF-15 inhibitor in accordance with the invention is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the antibody or antigen-binding portion thereof comprises a heavy chain variable domain which comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 3, a CDR2 region comprising the amino acid sequence of SEQ ID NO: 4 and a CDR3 region comprising the amino acid sequence of SEQ ID NO: 5, and wherein the antibody or antigen-binding portion thereof comprises a light chain variable domain which comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 6, a CDR2 region comprising the amino acid sequence ser-ala-ser and a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7.
  • the heavy chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 1 or a sequence 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical thereto
  • the light chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 2 or a sequence 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical thereto.
  • the antibody is a humanized antibody or an antigen-binding portion thereof.
  • the constant domain of the heavy chain of this monoclonal antibody or antigen-binding portion thereof may comprise the amino acid sequence of SEQ ID No: 29, or an amino acid sequence at least 85%, preferably at least 90%, more preferably at least 95% identical thereto, and the constant domain of the light chain of this monoclonal antibody or antigen-binding portion thereof may comprise the amino acid sequence of SEQ ID No: 32, or an amino acid sequence at least 85%, preferably at least 90%, more preferably at least 95% identical thereto.
  • the constant domain of the heavy chain of this monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 29, or an amino acid sequence at least 98%, preferably at least 99% identical thereto
  • the constant domain of the light chain of this monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 32, or an amino acid sequence at least 98%, preferably at least 99% identical thereto.
  • the constant domain of the heavy chain of this monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 29, and the constant domain of the light chain of this monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 32.
  • the heavy chain variable domain of this monoclonal antibody or antigen-binding portion thereof may comprise the amino acid sequence of SEQ ID No: 28, or an amino acid sequence at least 90%, preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto, and the light chain variable domain of this monoclonal antibody or antigen-binding portion thereof may comprise the amino acid sequence of SEQ ID No: 31, or an amino acid sequence at least 90%, preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto.
  • the heavy chain variable domain of this monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 28, and the light chain variable domain of this monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 31.
  • the heavy chain variable domain comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 3 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 4
  • the light chain variable domain comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 6 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 7.
  • the antibody may have CDR3 sequences as defined in any of the embodiments of the invention described above.
  • the antigen-binding portion is a single-domain antibody (also referred to as “NanobodyTM”).
  • the single-domain antibody comprises the CDR1, CDR2, and CDR3 amino acid sequences of SEQ ID NO: 3, SEQ ID NO: 4, and SEQ ID NO: 5, respectively.
  • the single-domain antibody comprises the CDR1, CDR2, and CDR3 amino acid sequences of SEQ ID NO: 6, ser-ala-ser, and SEQ ID NO: 7, respectively.
  • the single-domain antibody is a humanized antibody.
  • the antibodies capable of binding to human GDF-15 or the antigen-binding portions thereof have an equilibrium dissociation constant for human GDF-15 that is equal to or less than 100 nM, less than 20 nM, preferably less than 10 nM, more preferably less than 5 nM and most preferably between 0.1 nM and 2 nM.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof binds to the same human GDF-15 epitope as the antibody to human GDF-15 obtainable from the cell line B1-23 deposited with the Deutsche Sammlung für Mikroorganismen and Zellkulturen GmbH (DMSZ) under the accession No. DSM ACC3142.
  • DMSZ Deutsche Sammlung für Mikroorganismen and Zellkulturen GmbH
  • antibody binding to human GDF-15 in accordance with the present invention is preferably assessed by surface plasmon resonance measurements as a reference standard method, in accordance with the procedures described in Reference Example 1.
  • Binding to the same epitope on human GDF-15 can be assessed similarly by surface plasmon resonance competitive binding experiments of the antibody to human GDF-15 obtainable from the cell line B1-23 and the antibody that is expected to bind to the same human GDF-15 epitope as the antibody to human GDF-15 obtainable from the cell line B1-23.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 39 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto, and the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 40 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 41 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto, and the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 42 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto.
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 41 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 43 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto, and the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 44 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 45 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto, and the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 46 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto.
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 45 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 47 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto, and the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 48 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 49 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto, and the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 50 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 51 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto, and the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 52 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical thereto.
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 51 or a sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody or antigen-binding portion thereof, which is capable to compete with any one of the antibodies capable of binding to human GDF-15 referred to herein for binding to human GDF-15, preferably for binding to recombinant human GDF-15.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a humanized monoclonal antibody or an antigen-binding portion thereof.
  • humanized monoclonal anti-human-GDF-15 antibodies of the invention or antigen-binding portions thereof can be generated in accordance with techniques known in the art, as described above.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the binding is binding to a conformational or discontinuous epitope on human GDF-15 comprised by the amino acid sequences of SEQ ID No: 25 and SEQ ID No: 26.
  • the antibody or antigen-binding portion thereof is an antibody or antigen-binding portion thereof as defined by the sequences of any one of the above embodiments.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof can be linked to a drug.
  • the drug can be a known anticancer agent and/or an immune-stimulatory molecule.
  • known anticancer agents include alkylating agents such as cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, and ifosfamide; anti-metabolites such as azathioprine and mercaptopurine; alkaloids such as vinca alkaloids (e.g. vincristine, vinblastine, vinorelbine, and vindesine), taxanes (e.g.
  • topoisomerase inhibitors such as camptothecins (e.g. irinotecan and topotecan); cytotoxic antibiotics such as actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and mitomycin; and radioisotopes.
  • camptothecins e.g. irinotecan and topotecan
  • cytotoxic antibiotics such as actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and mitomycin; and radioisotopes.
  • the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is modified by an amino acid tag.
  • tags include Polyhistidin (His-) tags, FLAG-tag, Hemagglutinin (HA) tag, glycoprotein D (gD) tag, and c-myc tag. Tags may be used for various purposes. For instance, they may be used to assist purification of the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof. Preferably, such tags are present at the C-terminus or N-terminus of the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof.
  • Cancer cells harbor genomic mutations which give rise to cancer cell antigens that are specific to the cancer cells and different from the antigens of non-cancerous cells. Therefore, an intact immune system which is not inhibited should recognize these cancer cell antigens, such that an immune response against these antigens is elicited.
  • Most cancers have developed immune tolerance mechanisms against these antigens.
  • One class of mechanisms by which cancer cells achieve such immune tolerance is the utilization of immune checkpoints.
  • An “immune checkpoint” as used herein generally means an immunological mechanism by which an immune response can be inhibited.
  • an immune checkpoint is a mechanism which is characterized in that a molecule of the immune system (or a group of molecules of the immune system) inhibits the immune response by inhibiting the activation of cells of the immune system.
  • a molecule of the immune system or a group of molecules of the immune system
  • Such molecule (or group of molecules) of the immune system which inhibits (inhibit) the immune response by inhibiting the activation of cells of the immune system is (are) also known as checkpoint molecule(s).
  • an “immune checkpoint blocker” is a molecule which is capable of blocking an immune checkpoint. While it is understood that an hGDF-15 inhibitor as used according to the invention has effects on the immune system including effects on CD8+ T cells, the term “immune checkpoint blocker” as used herein does not refer to an hGDF-15 inhibitor but means a molecule which is different from an hGDF-15 inhibitor.
  • immune checkpoint blockers which are known to date are inhibitors of immune checkpoint molecules such as inhibitors of human PD-1 and inhibitors of human PD-L1. Further immune checkpoint blockers are anti-LAG-3, anti-B7H3, anti-TIM3, anti-VISTA, anti-TIGIT, anti-KIR, anti-CD27, anti-CD137 as well as inhibitors of IDO. Therefore, as used in accordance with the present invention, a preferred form of an immune checkpoint blocker is an inhibitor of an immune checkpoint molecule.
  • an immune checkpoint blocker can be an activator of a co-stimulating signal which overrides the immune checkpoint.
  • Methods to measure the potency of immune checkpoint blockers include in vitro binding assays, primary T cell-based cytokine release assays, and in vivo model systems. Additionally, Promega has now developed a commercially available bioluminescent reporter system for PD-1/PD-L1, which is, for instance referred to in Mei Cong, Ph.D. et al.: Advertorial: Novel Bioassay to Assess PD-1/PD-L1 Therapeutic Antibodies in Development for Immunotherapy Bioluminescent Reporter-Based PD-1/PD-L1 Blockade Bioassay.
  • Preferred immune checkpoint blockers are inhibitors of human PD-1 and inhibitors of human PD-L1. In one preferred embodiment in accordance with all of the embodiments of the invention, the immune checkpoint blocker is not an inhibitor of human CTLA4.
  • an “inhibitor of human PD-1” can be any molecule which is capable of specifically inhibiting the function of human PD-1.
  • Non-limiting examples of such molecules are antibodies capable of binding to human PD-1 and DARPins (Designed Ankyrin Repeat Proteins) capable of binding to human PD-1.
  • the inhibitor of PD-1 to be used in accordance with the invention is an antibody capable of binding to human PD-1, more preferably a monoclonal antibody capable of binding to human PD-1.
  • the monoclonal antibody capable of binding to human PD-1 is selected from the group consisting of nivolumab, pembrolizumab, pidilizumab and AMP-224.
  • an “inhibitor of human PD-L1” can be any molecule which is capable of specifically inhibiting the function of human PD-L1.
  • Non-limiting examples of such molecules are antibodies capable of binding to human PD-L1 and DARPins (Designed Ankyrin Repeat Proteins) capable of binding to human PD-L1.
  • the inhibitor of human PD-L1 to be used in accordance with the invention is an antibody capable of binding to human PD-L1, more preferably a monoclonal antibody capable of binding to human PD-L1.
  • the monoclonal antibody capable of binding to human PD-L1 is selected from the group consisting of BMS-936559, MPDL3280A, MED14736, and MSB0010718C.
  • the methods used in the present invention are performed in accordance with procedures known in the art, e.g. the procedures described in Sambrook et al. (“Molecular Cloning: A Laboratory Manual.”, 2 nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. 1989), Ausubel et al. (“Current Protocols in Molecular Biology.” Greene Publishing Associates and Wiley Interscience; New York 1992), and Harlow and Lane (“Antibodies: A Laboratory Manual” Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. 1988), all of which are incorporated herein by reference.
  • Binding of antibodies to their respective target proteins can be assessed by methods known in the art.
  • the binding of monoclonal antibodies to their respective targets is preferably assessed by surface plasmon resonance measurements. These measurements are preferably carried out by using a Biorad ProteOn XPR36 system and Biorad GLC sensor chips, as exemplified for anti-human GDF-15 mAb-B1-23 in Reference Example 1.
  • Sequence Alignments of sequences according to the invention are performed by using the BLAST algorithm (see Altschul et al. (1990) “Basic local alignment search tool.” Journal of Molecular Biology 215. p. 403-410; Altschul et al.: (1997) Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25:3389-3402).
  • the following parameters are used: Max target sequences 10; Word size 3; BLOSUM 62 matrix; gap costs: existence 11, extension 1; conditional compositional score matrix adjustment.
  • terms such as “identity” or “identical” refer to the identity value obtained by using the BLAST algorithm.
  • Monoclonal antibodies according to the invention can be produced by any method known in the art, including but not limited to the methods referred to in Siegel D L (“Recombinant monoclonal antibody technology.” Transfus Clin Biol. 2002 January; 9(1):15-22).
  • an antibody according to the invention is produced by the hybridoma cell line B1-23 deposited with the Deutsche Sammlung für Mikroorganismen and Zellkulturen GmbH (DSMZ) at Inhoffenstra ⁇ e 7B, 38124 Braunschweig, Germany, under the accession No. DSM ACC3142 under the Budapest treaty. The deposit was filed on Sep. 29, 2011.
  • Cell proliferation can be measured by suitable methods known in the art, including (but not limited to) visual microscopy, metabolic assays such as those which measure mitochondrial redox potential (e.g. MTT (344,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay; Resazurin staining which is also known as Alamar Blue® assay), staining of known endogenous proliferation biomarkers (e.g. Ki-67), and methods measuring cellular DNA synthesis (e.g. BrdU and [ 3 H]-Thymidine incorporation assays).
  • MTT 344,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • Resazurin staining which is also known as Alamar Blue® assay
  • staining of known endogenous proliferation biomarkers e.g. Ki-67
  • methods measuring cellular DNA synthesis e.g. BrdU and [ 3
  • hGDF-15 levels of human GDF-15 can be measured by any method known in the art, including measurements of hGDF-15 protein levels by methods including (but not limited to) mass spectrometry for proteins or peptides derived from human GDF-15, Western Blotting using antibodies specific to human GDF-15, flow cytometry using antibodies specific to human GDF-15, strip tests using antibodies specific to human GDF-15, or immunocytochemistry using antibodies specific to human GDF-15.
  • a preferred method of measuring hGDF-15 serum levels is a measurement of hGDF-15 serum levels by Enzyme-Linked Immunosorbent Assay (ELISA) by using antibodies to GDF-15. Such ELISA methods are exemplified in Example 1.
  • hGDF-15 serum levels may be determined by known electrochemiluminesence immunoassays using antibodies to GDF-15. For instance, the Roche Elecsys® technology can be used for such electrochemiluminesence immunoassays.
  • compositions in accordance with the present invention are prepared in accordance with known standards for the preparation of pharmaceutical compositions.
  • compositions are prepared in a way that they can be stored and administered appropriately, e.g. by using pharmaceutically acceptable components such as carriers, excipients or stabilizers.
  • Such pharmaceutically acceptable components are not toxic in the amounts used when administering the pharmaceutical composition to a patient.
  • the pharmaceutical acceptable components added to the pharmaceutical compositions may depend on the chemical nature of the inhibitors present in the composition (e.g. depend on whether the inhibitors are antibodies, si RNA hairpin constructs or short interfering RNAs), the particular intended use of the pharmaceutical compositions and the route of administration.
  • the pharmaceutically acceptable components used in connection with the present invention are used in accordance with knowledge available in the art, e.g. from Remington's Pharmaceutical Sciences, Ed. AR Gennaro, 20th edition, 2000, Williams & Wilkins, Pa., USA.
  • the present invention relates to the hGDF-15 inhibitors for the uses as defined above.
  • the present invention also relates to corresponding therapeutic methods.
  • the invention relates to a method for increasing the percentage of CD8 + T-cells in a solid cancer in a human patient, the method comprising the step of administering an hGDF-15 inhibitor to the human patient.
  • the invention in another embodiment, relates to a method of treating a solid cancer by an immune checkpoint blocker in a human patient, the method comprising a step of administering an hGDF-15 inhibitor to the human patient and a step of administering the immune checkpoint blocker to the human patient.
  • hGDF-15 inhibitors for use, kits, compositions, or compositions for use, the hGDF-15 inhibitor is the sole ingredient which is pharmaceutically active against cancer.
  • hGDF-15 inhibitors for use, kits, compositions, or compositions for use, the hGDF-15 inhibitor and the immune checkpoint blocker are the sole ingredients which are pharmaceutically active against cancer.
  • hGDF-15 inhibitors for use is used in combination with one or more further ingredients pharmaceutically active against cancer.
  • the one or more further ingredients pharmaceutically active against cancer is a known anticancer agent and/or an immune-stimulatory molecule.
  • anticancer agents include but are not limited to alkylating agents such as cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, and ifosfamide; anti-metabolites such as azathioprine and mercaptopurine; alkaloids such as vinca alkaloids (e.g. vincristine, vinblastine, vinorelbine, and vindesine), taxanes (e.g. paclitaxel, docetaxel) etoposide and teniposide; topoisomerase inhibitors such as camptothecins (e.g.
  • cytotoxic antibiotics such as actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and mitomycin; and radioisotopes.
  • Immune-stimulatory molecules include anti-LAG-3, anti-B7H3, anti-TIM3, anti-VISTA, anti-TIGIT, anti-KIR, anti-CD27, anti-CD137, anti-Ox40, anti-4-1BB, anti-GITR, anti-CD28, anti-CD40 or IDO-Inhibitors.
  • other antibody treatments like anti-Her2, anti-EGFR, anti-Claudin, or their glyco-optimized successors are also particularly preferred as they will benefit from a combination with the hGDF-15 inhibitor, e.g. due to enhanced immune cell infiltration in the solid cancer caused by the hGDF-15 inhibitor.
  • tumor-reactive T cells or dendritic cells are also particularly preferred as they will benefit from a combination with the hGDF-15 inhibitor.
  • tumor-reactive T cells or dendritic cells are also particularly preferred as they will benefit from a combination with the hGDF-15 inhibitor.
  • the following treatments are also particularly preferred as they will synergize with the hGDF-15 inhibitor:
  • the present invention encompasses combinations of an hGDF-15 inhibitor and an immune checkpoint blocker for use in a method of treating a solid cancer in a human patient, wherein the hGDF-15 inhibitor and the immune checkpoint blocker are to be administered to the human patient.
  • These combinations and their preferred embodiments are as defined above.
  • the combination of the hGDF-15 inhibitor and the immune checkpoint blocker may either be administered together or separately.
  • administration of the hGDF-15 inhibitor is to be started prior to the start of administration of the immune checkpoint blocker.
  • This setting advantageously allows to increase the percentage of T-cells, and in particular the percentage of CD8 + T cells in the solid cancer, such that a subsequent treatment with the immune checkpoint blocker can be more effective due to the increased starting percentage of CD8 + T cells in the solid cancer.
  • the present invention also provides a kit comprising an hGDF-15 inhibitor and at least one immune checkpoint blocker, as defined above.
  • the hGDF-15 inhibitor and one or more or all of the immune checkpoint blockers can be contained in separate containers or in a single container.
  • a container as used can be any type of container that is suitable to store the hGDF-15 inhibitor and/or the at least one immune checkpoint blocker.
  • Non-limiting examples of such containers are vials and pre-filled syringes.
  • the kit may contain further therapeutic agents.
  • the kit may contain one or more further ingredients pharmaceutically active against cancer.
  • the one or more further ingredients pharmaceutically active against cancer can be as defined above.
  • Such further ingredients pharmaceutically active against cancer may be used in the methods of the invention together with the hGDF-15 inhibitor and the at least one immune checkpoint blocker.
  • kits according to the invention further comprises instructions for use.
  • amino acid sequences referred to in the present application are as follows (in an N-terminal to C-terminal order; represented in the one-letter amino acid code):
  • SEQ ID No: 1 (Region of the Heavy Chain Variable Domain comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region from the Polypeptide Sequence of monoclonal anti-human GDF-15 mAb-B1-23): QVKLQQSGPGILQSSQTLSLTCSFSGFSLSTSGMGVSWIRQPSGKGLEWLAHIYWDDDKRYNPTLKSRLTISK DPSRNQVFLKITSVDTADTATYYC SEQ ID No: 2 (Region of the Light Chain Variable Domain comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region from the Polypeptide Sequence of monoclonal anti-human GDF-15 mAb-B1-23): DIVLTQSPKFMSTSVGDRVSVTCKASQNVGTNVAWFLQKPGQSPKALIYSASYRYSGVPDRFTGSGSGTDFT LTISNVQSEDLAEYFC SEQ ID No: 3 (
  • nucleic acid sequences referred to in the present application are as follows (in a 5′ to 3′ order; represented in accordance with the standard nucleic acid code):
  • SEQ ID No: 21 (DNA nucleotide sequence encoding the amino acid sequence defined in SEQ ID No: 1): CAAGTGAAGCTGCAGCAGTCAGGCCCTGGGATATTGCAGTCCTCCCAGAC CCTCAGTCTGACTTGTTCTTTCTCTGGGTTTTCACTGAGTACTTCTGGTA TGGGTGTGAGCTGGATTCGTCAGCCTTCAGGAAAGGGTCTGGAGTGGCTG GCACACATTTACTGGGATGATGACAAGCGCTATAACCCAACCCTGAAGAG CCGGCTCACAATCTCCAAGGATCCCTCCAGAAACCAGGTATTCCTCAAGA TCACCAGTGTGGACACTGCAGATACTGCCACATACTACTGT SEQ ID No: 22 (DNA nucleotide sequence encoding the amino acid sequence defined in SEQ ID No: 2): GACATTGTGCTCACCCAGTCTCCAAAATTCATGTCCACATCAGTAGGAGA CAGGGTCAGCGTCACCTGCAAGGCCAGTCAGAATGTGGGTACTAATGTGG CCTG
  • SEQ ID No: 27 amino acid sequence of the heavy chain of the H1L5 humanized B1-23 anti-GDF-15 antibody: QITLKESGPTLVKPTQTLTLTCTFSGFSLSTSGMGVSWIRQPPGKGLEWLAHIYWDDDKRYNPTLKSRLTITKD PSKNQVVLTMTNMDPVDTATYYCARSSYGAMDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGC LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKKVEP KSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKT KPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKN QVSLTCLVKGF
  • hGDF-15 inhibitor which can be used in the compositions, kits, methods and uses according to the invention.
  • This hGDF-15 inhibitor is a monoclonal antibody which is known from WO 2014/049087, which is incorporated herein by reference in its entirety:
  • the antibody B1-23 was generated in a GDF-15 knock out mouse.
  • Recombinant human GDF-15 (SEQ ID No: 8) was used as the immunogen.
  • the hybridoma cell line B1-23 producing mAb-B1-23 was deposited by the Julius-Maximilians-Universitat Würzburg, Sanderring 2, 97070 Würzburg, Germany, with the Deutsche Sammlung für Mikroorganismen and Zellkulturen GmbH (DMSZ) at Inhoffenstra ⁇ e 7B, 38124 Braunschweig, Germany, under the accession No. DSM ACC3142, in accordance with the Budapest Treaty. The deposit was filed on Sep. 29, 2011.
  • Kd dissociation constant
  • Binding of the monoclonal anti-human-GDF-15 antibody anti-human GDF-15 mAb-B1-23 according to the invention was measured by employing surface plasmon resonance measurements using a Biorad ProteOn XPR36 system and Biorad GLC sensor chips:
  • recombinant mature human GDF-15 protein was immobilized on flow cells 1 and 2. On one flow cell recombinant GDF-15 derived from Baculvirus-transfected insect cells (HighFive insect cells) and on the other recombinant protein derived from expression in E. coli was used.
  • the non-reacted coupling groups were then quenched by perfusion with 1M ethanolamine pH 8.5 and the biosensor was equilibrated by perfusing the chip with running buffer (10M HEPES, 150 mM NaCl, 3.4 mM EDTA, 0.005% Tween-20, pH 7.4, referred to as HBS150).
  • running buffer 10M HEPES, 150 mM NaCl, 3.4 mM EDTA, 0.005% Tween-20, pH 7.4, referred to as HBS150.
  • HBS150 running buffer
  • two flow cells were used, one empty with no protein coupled and one coupled with an non-physiological protein partner (human Interleukin-5), which was immobilized using the same coupling chemistry and the same coupling density.
  • anti-human GDF-15 mAb-B1-23 was dissolved in HBS150 and used in six different concentrations as analyte (concentration: 0.4, 0.8, 3, 12, 49 and 98 nM).
  • concentration: 0.4, 0.8, 3, 12, 49 and 98 nM concentration: 0.4, 0.8, 3, 12, 49 and 98 nM.
  • the analyte was perfused over the biosensor using the one-shot kinetics setup to avoid intermittent regeneration, all measurements were performed at 25° C. and using a flow rate of 100 ⁇ l min ⁇ 1 .
  • For processing the bulk face effect and unspecific binding to the sensor matrix was removed by subtracting the SPR data of the empty flow cell (flow cell 3) from all other SPR data.
  • the resulting sensogram was analyzed using the software ProteOn Manager version 3.0. For analysis of the binding kinetics a 1:1 Langmuir-type interaction was assumed.
  • the anti-human GDF-15 mAb-B1-23 shows no binding to human interleukin-5 and thus confirms the specificity of the interaction data and the anti-human GDF-15 mAb-B1-23.
  • the amino acid sequence of recombinant human GDF-15 (as expressed in Baculovirus-transfected insect cells) is:
  • the dissociation constant (Kd) of 790 pM was determined.
  • mAb B1-23 inhibits cancer cell proliferation in vitro, and that mAb B1-23 inhibits growth of tumors in vivo (WO2014/049087).
  • the protein sequence was translated into 13mer peptides with a shift of one amino acid.
  • the C- and N-termini were elongated by a neutral GSGS linker to avoid truncated peptides (bold letters).
  • Blocking buffer Rockland blocking buffer MB-070
  • Monoclonal mouse antibody GDF-15 (1 ⁇ g/ ⁇ l): Staining in incubation buffer for 16 h at 4° C. at a dilution of 1:100 and slight shaking at 500 rpm
  • Control antibodies Monoclonal anti-HA (12CA5)-LL-Atto 680 (1:1000), monoclonal anti-FLAG(M2)-FluoProbes752 (1:1000); staining in incubation buffer for 1 h at RT
  • the peptide array with 10, 12 and 15mer B7H3-derived linear peptides was incubated with secondary goat anti-mouse IgG (H+L) IRDye680 antibody only at a dilution of 1:5000 for 1 h at room temperature to analyze background interactions of the secondary antibody.
  • the PEPperCHIP® was washed 2 ⁇ 1 min with standard buffer, rinsed with dist. water and dried in a stream of air.
  • the peptide microarray was incubated overnight at 4° C. with monoclonal mouse antibody GDF-15 at a dilution of 1:100. Repeated washing in standard buffer (2 ⁇ 1 min) was followed by incubation for 30 min with the secondary antibody at a dilution of 1:5000 at room temperature. After 2 ⁇ 10 sec. washing in standard buffer and short rinsing with dist. water, the PEPperCHIP® was dried in a stream of air. Read-out was done with Odyssey Imaging System at a resolution of 21 ⁇ m and green/red intensities of 7/7 before and after staining of control peptides by anti-HA and anti-FLAG(M2) antibodies.
  • the epitope of recombinant human GDF-15 which binds to the antibody B1-23 was identified by means of the epitope excision method and epitope extraction method (Suckau et al. Proc Natl Acad Sci USA. 1990 December; 87(24): 9848-9852; R. Stefanescu et al., Eur. J. Mass Spectrom. 13, 69-75 (2007)).
  • the antibody B1-23 was added to NHS-activated 6-aminohexanoic acid coupled sepharose.
  • the sepharose-coupled antibody B1-23 was then loaded into a 0.8 ml microcolumn and washed with blocking and washing buffers.
  • Recombinant human GDF-15 was digested with trypsin for 2 h at 37° C. (in solution), resulting in different peptides, according to the trypsin cleavage sites in the protein. After complete digestion, the peptides were loaded on the affinity column containing the immobilized antibody B1-23. Unbound as well as potentially bound peptides of GDF-15 were used for mass spectrometry analysis. An identification of peptides by means of mass spectrometry was not possible. This was a further indicator that the binding region of GDF-15 in the immune complex B1-23 comprises a discontinuous or conformational epitope.
  • the digested peptides should bind its interaction partner, unless there was a trypsin cleavage site in the epitope peptide.
  • a discontinuous or conformational epitope could be confirmed by the epitope excision method described in the following part.
  • the immobilized antibody B1-23 on the affinity column was then incubated with recombinant GDF-15 for 2 h.
  • the formed immune complex on the affinity column was then incubated with trypsin for 2 h at 37° C.
  • the cleavage resulted in different peptides derived from the recombinant GDF-15.
  • the immobilized antibody itself is proteolytically stable.
  • the resulting peptides of the digested GDF-15 protein, which were shielded by the antibody and thus protected from proteolytic cleavage, were eluted under acidic conditions (TFA, pH2), collected and identified by mass spectrometry.
  • the part of human GDF-15, which binds the antibody B1-23, comprises a discontinuous or conformational epitope.
  • Mass spectrometry identified 2 peptides in the GDF-15 protein, which are responsible for the formation of the immune complex. These peptides are restricted to the positions 40-55 (EVQVTMCIGACPSQFR) and 94-114 (TDTGVSLQTYDDLLAKDCHCI) in the GDF-15 amino acid sequence. Thus, these two peptides comprise an epitope of the GDF-15 protein that binds to the antibody B1-23.
  • hGDF-15 Serum Levels correlate with poor treatment response at a time point of four months after the start of the treatment with pembrolizumab.
  • the present inventors set out to investigate whether cancer patients receiving immune checkpoint blockers could benefit from an inhibition of hGDF-15.
  • sera from melanoma patients which had received a prior treatment with Ipilimumab (a monoclonal anti-CTLA4 antibody) and received a treatment with Pembrolizumab (a monoclonal anti-PD-1 antibody) in a clinical study, were analyzed for hGDF-15 serum levels.
  • the obtained hGDF-15 serum levels were then correlated with the patients' responses. Sera were taken from the patients prior to the treatment with Pembrolizumab.
  • Eligible patients were aged 18 years or older and had histologically or cytologically confirmed unresectable stage III or stage IV melanoma not amenable to local therapy; confirmed disease progression within 24 weeks of the last ipilimumab dose (minimum two doses, 3 mg/kg once every 3 weeks); previous BRAF or MEK inhibitor therapy or both (if BRAFV600 mutant-positive); resolution or improvement of ipilimumab-related adverse events to grade 0-1 and prednisone dose 10 mg/day or less for at least 2 weeks before the first dose of study drug; Eastern Cooperative Oncology Group (ECOG) performance status 0 or 1; measurable disease per Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST v1.1); and values within the prespecified range for absolute neutrophil count ( ⁇ 1500 cells per mL), platelets ( ⁇ 100 000 cells per mL), haemoglobin ( ⁇ 90 g/L), serum creatinine ( ⁇ 1.5 upper limit of normal [ULN]), serum
  • ipilimumab a monoclonal anti-CTLA4 antibody
  • Pembrolizumab a monoclonal anti-PD-1 antibody
  • FIG. 1 shows the GDF-15 serum levels for responders and non-responders to the treatment regimen. As can be seen from the Figure, most of the non-responders have higher GDF-15 serum levels than all of the responders.
  • FIG. 2 shows the numbers of responders and non-responders in the patients having hGDF-15 serum levels of ⁇ 1.8 ng/ml, 1.8-4.2 ng/ml, and >4.2 ng/ml, respectively.
  • the data analysis was based on a data file containing data from samples from 35 patients containing the columns (variables) Sample designation, GDF-15 (ng/ml), responder/non-responder, days (to death or censoring), and Ongoing (an index variable for ongoing life).
  • the responder/non-responder classification of these data was made at a time point of four months after the start of the treatment with pembrolizumab. As some serum samples had only been obtained shortly before the analysis, response could only by assessed in 29 patients. One partial responder (>30% reduction in tumor size) was rated as responder. For LDH determination, 4 samples had to be excluded due to hemolysis.
  • Tables 1-2 show the results from the models with GDF-15 as continuous predictor.
  • the hazard for death significantly increased for higher concentrations of GDF-15 (HR>1, Table 1) whereas the probability of response to treatment significantly decreased, as indicated by the odds ratio (OR) (OR ⁇ 1, Table 2).
  • FIG. 3 shows the corresponding data on responders/non-responders as well as the probability of response to treatment predicted by the model.
  • Table 3 shows the result from the Cox proportional hazards model with the group based on GDF-15 as categorical predictor.
  • the group with GDF-15 ⁇ 1.8 ng/ml is used as reference group (not shown in the Table).
  • the two hazard ratios in Table 3 represent the comparison of the group with GDF-15 between 1.8 and 4.2 and the group with GDF-15 >4.2 with the reference group.
  • the hazard for death is increased in both of these groups (compared to the reference group), but to a larger extent in the group with GDF-15 >4.2.
  • FIG. 4 shows the Kaplan-Meier curves for survival in the three groups.
  • Table 1 shows the Hazard ratio (HR) estimates from the Cox proportional hazards model with overall survival (time to death) as outcome variable and GDF-15 as continuous predictor.
  • the analysis included samples from 35 patients.
  • Table 2 shows the Odds ratio (OR) estimates from the Generalized Linear Model with response to treatment (responder 1) as outcome variable and GDF-15 as continuous predictor.
  • the analysis included samples from 29 patients.
  • Table 3 shows Hazard ratio (HR) estimates from the Cox proportional hazards model with overall survival (time to death) as outcome variable and the group based on GDF-15 as categorical predictor.
  • the analysis included samples from 35 patients.
  • Table 4 shows the number of deaths and responders (responder1) in the three groups defined by the GDF-15 ( ⁇ 1.8, 1.8-4.2, >4.2 ng/ml).
  • LDH may be interpreted as reflecting high tumor burden or tumor aggressiveness (Zhang, J., Yao, Y.-H., Li, B.-G., Yang, Q., Zhang, P.-Y., and Wang, H.-T. (2015). Prognostic value of pretreatment serum lactate dehydrogenase level in patients with solid tumors: a systematic review and meta-analysis. Scientific Reports 5, 9800). As serum LDH levels have been incorporated into the staging scheme for melanoma, this parameter is routinely measured during clinical diagnostics by the university reference laboratory.
  • Table 7 is analogue to Table 2, except that LDH was used as continuous predictor of response to treatment (responder1) instead of GDF-15.
  • FIGS. 5A-5B show the corresponding data on responders/non-responders as well as the probability of response to treatment predicted by the model.
  • GDF-15 is the better predictor of response to treatment (responder1) than LDH
  • two additional models were fitted: a model containing both markers as predictors (which automatically only includes patients with measurements on both markers), and a model with GDF-15 as the only predictor but also only using the patients with a measurement of LDH.
  • AIC Akaike's information criterion
  • FIG. 5A shows the probability of response to treatment (responder 1) as predicted by the Generalized Linear Model model using LDH as continuos predictor. Circles show the data, the curve shows the model. The vertical line indicates the LDH concentration where the probability of treatment response is 0.5.
  • the patient cohort was identical. However, reliable determination of LDH levels failed in four patients due to hemolysis.
  • FIG. 5B shows a graphical representation of responders and non-responders and their respective hGDF-15 and LDH levels. When cut-off values are selected to cover all responders, testing based on GDF-15 allows for identification of 6 (out of 9) non-responders whereas analyses based on LDH levels can only discriminate 4 (out of 9) non-responders. For LDH testing, 4 hemolytic samples had to be excluded which causes loss of data.
  • Example 1 showed that the likelihood of a response to the treatment significantly decreases with increasing hGDF-15 levels in the patient sera.
  • Example 1 The results of Example 1 suggest that hGDF-15 acts to negatively affect the patients' responses to the treatment with immune checkpoint blockers.
  • an inhibitor of hGDF-15 will be useful to inhibit the negative effects of hGDF-15 on the patients' responses to the treatment with immune checkpoint blockers, and to improve the patients' responses to the treatment with immune checkpoint blockers not only in melanoma, but in all of the solid cancers referred to herein.
  • TILs Tumor Infiltrating Lymphocytes
  • hGDF-15 In order to identify a mechanism of hGDF-15 that contributes to the negative effect of hGDF-15 on the patients' responses, brain metastases from different solid tumors were analyzed for the expression of hGDF-15 and for the presence of cells of the immune system:
  • FFPE Formalin-fixed and paraffin-embedded
  • Immunohistochemistry for all antibodies was performed using 3 ⁇ m thick slides and standard protocols on the automated IHC staining system Discovery XT (RocheNentana, Arlington, Ariz., USA). The following antibodies were used: anti GDF-15 (HPA011191, dilution 1:50, Sigma/Atlas, protocol #730), CD3 (clone A0452, dilution 1:500, DAKO, Glostrup, Denmark), CD8 (clone C8/144B, dilution 1:100, DAKO, Glostrup, Denmark), PD-1 (clone NAT105; dilution 1:50; Abcam, Cambridge, United Kingdom), PD-L1 (E1L3N; dilution 1:200; Cell Signaling, Boston, U.S.A.), FOXP3 (clone 236A/E7; dilution 1:100; eBioscience, San Diego, U.S.A.). Slides were counterstained with hematoxylin and mounted
  • FIG. 6 shows exemplary tissue sections from melanoma brain metastases having high no (upper panel) or high (lower panel) GDF-15 immunoreactivity, which were stained by immunohistochemistry for GDF-15 and for the T-cell marker proteins CD3 and CD8, respectively, as indicated in the Figure.
  • the numerous infiltrating immune cells are seen as dark spots.
  • the scarce infiltrating immune cells are depicted by arrows (CD3 and CD8-positive cells are indicated by arrows).
  • FIG. 8 shows a plot of the GDF-15 score against the percentage of CD8 + and CD3 + T cells, respectively, in 168 (for CD3) or, respectively, 169 (for CD8) brain metastases from different tumor entities (melanoma, CRC, RCC, breast cancer, NSCLC and SCLC). The plot was obtained as described above in the “statistical analyses” section. As indicated in FIG.
  • therapeutic inhibition of hGDF-15 can be used to increase the percentage of CD8 + T lymphocytes in solid tumors including tumor metastases.
  • This increase of CD8 + T lymphocytes in the solid tumors can be used for therapy of the solid tumors.
  • a particularly favorable therapeutic combination is the combination of an hGDF-15 inhibitor with an immune checkpoint blocker.
  • An advantageous effect of this combination is that inhibition of hGDF-15 will increase the percentage of CD8 + T lymphocytes in the solid tumors and thereby lead to a synergistic therapeutic effect with immune checkpoint inhibition.
  • the invention can thus be applied to all of the solid tumors as referred to in the preferred embodiments.
  • the inventors next set out to determine how hGDF-15 affects the percentage of T cells in the solid tumors.
  • a step which is required for the invasion of T cells from the blood stream into the tumor tissue is that the T cells must first adhere to the endothelium before they can enter the tumor.
  • the inventors used a model system which measures the adhesion of T cells to Human Umbilical Vein Endothelial Cells (HUVEC):
  • Recombinant GDF-15 was obtained from Invigate GmbH, Jena, Germany.
  • FIGS. 9A-9D show analyses of several adhesion parameters, namely
  • FIG. 9C it was found that treatment of the T cells with hGDF-15 significantly decreases the adhesion to the endothelial cells, as reflected in the number of adhering cells per field of view. Similar results were obtained when analyzing adhesion by counting the numbers of rolling T cells ( FIG. 9A ). Furthermore, and consistent with the above results, it was found that treatment of the T cells with hGDF-15 significantly increases the rolling speed, indicating a decrease in the interaction time between the T cells and the endothelial cells, and also indicating a reduced adhesion between the T cells and the endothelial cells ( FIG. 9B ).
  • hGDF-15 acts both on the T cells and on the endothelial cells, but they also indicate that the main adhesion effect of hGDF-15 is an effect on the T cells.
  • hGDF-15 which is secreted by tumor cells, on T-cell adhesion could be inhibited with an hGDF-15 inhibitor.
  • the inventors used an hGDF-15-secreting melanoma cell line, UACC257:
  • Recombinant GDF-15 was obtained from Invigate GmbH, Jena, Germany.
  • FIG. 10A This Figure shows analyses of the number of rolling T cells per field of view per second.
  • the data were obtained from channel #1 (control T cells on unstimulated HUVEC as “neg. control”), channel #2 (control T cells on stimulated HUVEC as “pos. control”), channel #3 (“GDF-15”) channel #4 (“UACC 257”: T cells cultured in the supernatant of UACC 257 melanoma cells containing secreted GDF-15) and channel #5 (“UACC257+anti-hGDF-15”: T cells cultured in the supernatant of UACC 257 melanoma cells depleted from secreted GDF-15 with anti GDF-15 B1-23)
  • hGDF-15 inhibitors can be used to increase adhesion of T-cells including CD8+ cells to endothelial cells, e.g. in the treatment of solid cancers.
  • the above assay provides a simple in vitro system which can be used to determine whether a substance of interest is an hGDF-15 inhibitor.
  • a treatment with hGDF-15 inhibitors can be used to increase adhesion of T cells including CD8 + T cells to endothelial cells.
  • Such treatment will increase entry of T cells including CD8 + T cells from the blood stream into solid cancers.
  • the increased percentage of CD8 + T cells in solid cancers, which will result from such treatment with hGDF-15 inhibitors, is advantageous for, and can be used in, cancer therapy, e.g. cancer immunotherapy.
  • hGDF-15 inhibitors Since the entry of CD8 + T cells into solid cancers and the presence of these CD8 + T cells in the solid cancers is particularly advantageous for therapeutic approaches using immune checkpoint blockers, a particularly advantageous use of hGDF-15 inhibitors according to the invention is their use in combination with immune checkpoint blockers.
  • the flow/adhesion assay was conducted as described above in the present Example. T-cells were pre-incubated with 100 ng/ml GDF-15 for 1 hour or with 100 ng/ml GDF-15, which was pre-incubated with 10 ⁇ g/ml antibody for 1 hour.
  • the following Anti-GDF-15 antibodies were used: H1L5 (Humanized B1-23), 01G06 and 03G05 (Humanized Anti-GDF-15 Antibodies Engineered According to Sequences from WO 2014/100689).
  • hGDF-15 inhibitors can be used to increase adhesion of T-cells including CD8+ cells to endothelial cells, or the rolling of said T-cells including CD8+ cells.
  • hGDF-15 inhibitors will increase the percentage of CD8+ cells in solid cancers and can be used for the treatment of these cancers.
  • These hGDF-15 inhibitors may be—but are not limited to—any known anti-GDF-15 antibodies such as the antibodies H1L5 (Humanized B1-23), 01G06 and 03G05.
  • Example 4 Evaluation of Anti-Tumor Efficacy of Test Antibody in Combination with Adjuvant Immunization in Syngeneic MC38 tg hGDF-15+ Tumor-Bearing Mice
  • GDF-15 human growth & differentiation factor
  • mice On day 0, 9 week-old female C57BL/6J mice (provided by Charles River Laboratories, BP 0109, F 69592 L'Arbresle, Cedex) were anesthetized and subcutaneously inoculated with 2 ⁇ 10 5 colon MC38 tg hGDF-15 cells. Treatment with anti GDF-15 antibody (20 mg/kg of body weight, i.e. about 400 ⁇ g per mouse in 100 ⁇ l of phosphate-buffered saline with 0.5% bovine serum albumin) was initiated on day 0 (about 6 h after tumor cell inoculation) and repeated on days 3, 7, 10, 14, 17, and 21.
  • anti GDF-15 antibody (20 mg/kg of body weight, i.e. about 400 ⁇ g per mouse in 100 ⁇ l of phosphate-buffered saline with 0.5% bovine serum albumin) was initiated on day 0 (about 6 h after tumor cell inoculation) and repeated on days 3, 7, 10, 14, 17, and 21.
  • RNA Due to its structural similarity to double-stranded RNA, which is present in some viruses and stimulates TLR3, Poly-ICLC simulates an infection.
  • the agonistic anti-CD40 antibody provides an additional signal to antigen-presenting cells. “Licensing” of dendritic cells via stimulation of CD40 supports the activation of antigen-specific CD8 + T cells. Adjuvant treatment thus serves to induce tumor-specific immune cells in mice kept under specific pathogen-free conditions (Yadav Metal., Nature. 2014 Nov. 27; 515(7528):572-6).
  • This adjuvant treatment therefore represents a model system for a cancer immunotherapy which requires immune cells in the tumor, and in particular CD8 + T-cells in the tumor.
  • a model system which is suitable to further confirm that a treatment with an hGDF-15 inhibitor such as an anti-hGDF-15 antibody synergizes with cancer immunotherapy, including a cancer immunotherapy that requires CD8 + T-cells in the tumor.
  • mice per group were investigated:
  • Tumor size was measured 3 times per week by caliper-based measurement of tumor length and width.
  • mice For the surviving mice, the presence of tumors was determined by physical examination until day 57 past tumor inoculation. The results are shown in FIG. 11 .
  • results obtained in this model system further confirm that hGDF-15 inhibitors synergize with cancer immunotherapy, and in particular with cancer immunotherapy that requires the activation of immune cells such as CD8 + T-cells which then exert the cytotoxic activity in the tumor tissue.
  • results also further confirm that the increase in the percentage of CD8 + T-cells in the cancer, which is caused by the uses of hGDF-15 inhibitors according to the invention, can advantageously be used in cancer therapy.
  • the murine immune model system has very little time to build-up an antigen-specific CD8 + T cell response to a fast-growing cancer.
  • an adjuvant was used to further support the spontaneous immune response in the murine system.
  • antigen-specific T cells directed against cancer antigens are typically already present at diagnosis, i.e. priming of an immune response usually occurs even before the cancer is diagnosed.
  • These cancer antigen-specific CD8 + T cells already exist in humans but to a much lesser extent in the murine model system. Therefore, according to the invention, the uses of hGDF-15 inhibitors according to the invention will be even more effective in humans than in the present murine model system.
  • hGDF-15 inhibitors can effectively be used for the treatment of human cancer patients according to the invention, e.g. to increase in the percentage of CD8 + T-cells in a solid cancer, and they will synergize with other cancer immune therapies in humans, and in particular with cancer immune therapies which require CD8 + T-cells in the cancer, including cancer immunotherapies with immune checkpoint blockers such as anti-PD-1 and anti-PD-L1 antibodies.
  • Example 5 GDF-15 Serum Levels Define Survival of Melanoma Patients Treated with Anti PD-1
  • Example 2 The study in this Example was performed in order to further validate the results obtained in the study of Example 1, e.g. the finding that hGDF-15 influences the patients' response to immune checkpoint blockers, by an additional independent study.
  • CMMR Central Malignant Melanoma Registry
  • Prior treatment with ipilimumab was considered to have failed when patients had received a minimum of two doses, 3 mg/kg once every 3 weeks, but showed confirmed disease progression within 24 weeks of the last ipilimumab dose.
  • resolution or improvement of ipilimumab-related adverse events to grade 0-1 and prednisone dose 10 mg/day or less was demanded for at least 2 weeks before the first dose of study drug.
  • Eligible patients had Eastern Cooperative Oncology Group (ECOG) performance status 0 or 1; measurable disease per Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST v1.1); and values within the prespecified range for absolute neutrophil count ( ⁇ 1500 cells per mL), platelets ( ⁇ 100 000 cells per mL), haemoglobin ( ⁇ 90 g/L), serum creatinine ( ⁇ 1 ⁇ 5 upper limit of normal [ULN]), serum total bilirubin ( ⁇ 1 ⁇ 5 ULN or direct bilirubin ULN for patients with total bilirubin concentrations >1 ⁇ 5 ULN), aspartate and alanine aminotransferases ( ⁇ 2 ⁇ 5 ULN or ⁇ 5 ULN for patients with liver metastases), international normalised ratio or prothrombin time ( ⁇ 1 ⁇ 5 ULN if not using anticoagulants), and activated partial thromboplastin time ( ⁇ 1 ⁇ 5 ULN if not using anticoagulants). Patients had a washout period of
  • the data analysis was based on a data file containing data from samples from 99 patients containing the columns (variables) Sample designation, GDF-15 (ng/ml), days (to death or censoring), and Ongoing (an index variable for ongoing life).
  • the resulting Kaplan-Meier curves are shown in FIGS. 12 and 13 where censoring is indicated by vertical lines.
  • Table 9 patient survival data for patient groups having GDF-15 levels of ⁇ 1.5 ng/ml and ⁇ 1.5 ng/ml
  • Tables 10 and 11 total statistical comparisons of the patient groups having GDF-15 levels of ⁇ 1.5 ng/ml and ⁇ 1.5 ng/ml
  • Example 1 The above statistical results of this Example further confirmed the results of Example 1. For instance, it was confirmed that the likelihood of a response to the treatment, as indicated by the survival of the patients, significantly decreases with increasing hGDF-15 levels in the patient sera.
  • Table 12 shows that the survival between the two patient groups having GDF-15 levels of ⁇ 1.5 ng/ml and ⁇ 1.5 ng/ml, respectively, was significantly different, as evidenced by a significance level of 0.004.
  • Table 9 demonstrates that a higher percentage of patients (82.3%) survived in the group having GDF-15 levels of ⁇ 1.5 ng/ml
  • Tables 10 and 11 and FIGS. 12 and 13 demonstrate that for patients having GDF-15 levels of ⁇ 1.5 ng/ml, survival times were remarkably longer than in patients having GDF-15 levels of ⁇ 1.5 ng/ml.
  • hGDF-15 acts to negatively affect the patients' responses to the treatment with immune checkpoint blockers.
  • an inhibitor of hGDF-15 will be useful to inhibit the negative effects of hGDF-15 on the patients' responses to the treatment with immune checkpoint blockers, and to improve the patients' responses to the treatment with immune checkpoint blockers not only in melanoma, but in all of the solid cancers referred to herein.
  • Example 6 In Human Non-Small Cell Lung Cancer (NSCLC) Patients Treated with an Anti-PD1 Antibody, Median hGDF-15 Serum Levels in Patients with Progressive Disease are Higher than in Patients Showing a Partial Response
  • NSCLC Human Non-Small Cell Lung Cancer
  • Example 2 was performed in order to further validate the results obtained in the study of Example 1, e.g. the finding that hGDF-15 influences the patients' response to immune checkpoint blockers, in an additional independent study in a different solid cancer.
  • NSCLC patients were treated with anti-PD1 antibodies in accordance with the approved drug label of the anti-PD1 antibodies.
  • Serum samples were taken from the patients prior to the treatment with the anti-PD1 antibodies.
  • hGDF-15 serum levels in the serum samples were analyzed by Enzyme-Linked Immunosorbent Assay (ELISA), as described in Example 1.
  • ELISA Enzyme-Linked Immunosorbent Assay
  • the median hGDF-15 serum level in the patients showing a partial response was 0.55 ng/ml
  • the median hGDF-15 serum level in the patients showing progressive disease was 1.56 ng/ml.
  • the median hGDF-15 serum level in the patients showing a progressive disease was about 2.8-fold higher than in the patients showing a partial response.
  • results of this Example further confirm that hGDF-15 levels negatively correlate with the patients' response to immune checkpoint blockers.
  • the results of this Example also further confirm that hGDF-15 acts to negatively affect the patients' responses to the treatment with immune checkpoint blockers such as PD-1.
  • an inhibitor of hGDF-15 will be useful to inhibit the negative effects of hGDF-15 on the patients' responses to the treatment with immune checkpoint blockers, and to improve the patients' responses to the treatment with immune checkpoint blockers not only in melanoma, but also in lung cancers such as NSCLC and in all of the other solid cancers referred to herein.
  • Example 7 hGDF-15 Serum Levels do not Significantly Correlate with the Mutational Burden of the Tumors
  • the mutational burden is a known positive prognostic factor for a response of cancer patients to immune checkpoint blockers.
  • cancer cells harbor genomic mutations which give rise to cancer cell antigens that are specific to the cancer cells and different from the antigens of non-cancerous cells.
  • a high mutational burden leads to a high number of such cancer cell-specific antigens.
  • the stimulation of the immune response by immune checkpoint blockers is considered to be particularly effective, because more cancer cell-specific antigens are available as target antigens for the immune response.
  • hGDF-15 is not merely a surrogate marker for the mutational burden of the tumors
  • a treatment with hGDF-15 inhibitors acts via a mechanism that is independent from the mutational burden of the tumors
  • hGDF-15 mRNA levels in cancer samples from cancer patients were plotted against the number of somatic mutations which were identified in the cancers.
  • the somatic mutations were determined by use of exome sequencing.
  • the data were analyzed by using the UZH webtool from the University Hospital Zurich (Cheng P F et al.: Data mining The Cancer Genome Atlas in the era of precision cancer medicine. Swiss Med Wkly. 2015 Sep. 16; 145:w14183.) The results are shown in FIGS.
  • FIG. 14A shows a plot for cancer patient data obtained from the Cancer Genome Atlas (TGCA) considering only patients with high-grade malignant melanoma (the Cancer Genome Atlas is described in the reference of Cheng P F et al.: Data mining The Cancer Genome Atlas in the era of precision cancer medicine. Swiss Med Wkly. 2015 Sep. 16; 145:w14183).
  • GDF-15 expression was evaluated by normalization using the RSEM (“RNA Seq by expectation maximization”) software package (Li B and Dewey C N: RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011 Aug. 4; 12:323. doi: 10.1186/1471-2105-12-323).
  • FIG. 14B shows a plot for cancer patient data from 40 additional metastatic malignant melanoma patients from the University Hospital Zurich, which were separately analyzed.
  • FIGS. 14A and 14B show a p value of 0.5, indicating that there is no significant correlation between the mutational burden in the cancers and the levels of hGDF-15.
  • Example 8 CD8 + T-Cell Infiltration in Wild-Type Tumors or Human GDF-15 (Over)Expressing Tumors
  • hGDF-15 decreases the percentage of CD8 + T cells in solid cancers
  • hGDF-15 inhibitors such as anti-GDF-15 antibodies can be used to increase the percentage of CD8 + T-cells in a solid cancer in a human patient.
  • compositions and the kits according to the present invention may be industrially manufactured and sold as products for the claimed methods and uses (e.g. for treating a cancer as defined herein), in accordance with known standards for the manufacture of pharmaceutical products. Accordingly, the present invention is industrially applicable.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Organic Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Endocrinology (AREA)
  • Mycology (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US15/765,176 2015-10-02 2016-09-30 Combination therapy using inhibitors of human growth and differentiation factor 15 (GDF-15) and immune checkpoint blockers Active 2038-10-06 US11464856B2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GB1517531.8 2015-10-02
GBGB1517531.8A GB201517531D0 (en) 2015-10-02 2015-10-02 Combination therapy using inhibitors of human growth and differentiation factor 15 (GDF-15) and immune checkpoint blockers
GB1517531 2015-10-02
GB1607801 2016-04-29
GB1607801.6 2016-04-29
GBGB1607801.6A GB201607801D0 (en) 2016-04-29 2016-04-29 Combination therapy using inhibitors of human Growth and Differentiation Factor 15 (GDF-15) and immune checkpoint blockers
PCT/EP2016/073520 WO2017055613A2 (en) 2015-10-02 2016-09-30 Combination therapy using inhibitors of human growth and differentiation factor 15 (gdf-15) and immune checkpoint blockers

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/073520 A-371-Of-International WO2017055613A2 (en) 2015-10-02 2016-09-30 Combination therapy using inhibitors of human growth and differentiation factor 15 (gdf-15) and immune checkpoint blockers

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/823,383 Division US20230093412A1 (en) 2015-10-02 2022-08-30 Combination therapy using inhibitors of human growth and differentiation factor 15 (gdf-15) and immune checkpoint blockers

Publications (2)

Publication Number Publication Date
US20190160169A1 US20190160169A1 (en) 2019-05-30
US11464856B2 true US11464856B2 (en) 2022-10-11

Family

ID=57121227

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/765,176 Active 2038-10-06 US11464856B2 (en) 2015-10-02 2016-09-30 Combination therapy using inhibitors of human growth and differentiation factor 15 (GDF-15) and immune checkpoint blockers
US17/823,383 Pending US20230093412A1 (en) 2015-10-02 2022-08-30 Combination therapy using inhibitors of human growth and differentiation factor 15 (gdf-15) and immune checkpoint blockers

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/823,383 Pending US20230093412A1 (en) 2015-10-02 2022-08-30 Combination therapy using inhibitors of human growth and differentiation factor 15 (gdf-15) and immune checkpoint blockers

Country Status (22)

Country Link
US (2) US11464856B2 (zh)
EP (2) EP4218809A3 (zh)
JP (3) JP6925326B2 (zh)
KR (1) KR20180054868A (zh)
CN (2) CN117883572A (zh)
AU (2) AU2016333539B2 (zh)
BR (1) BR112018006218A2 (zh)
CA (1) CA3000293A1 (zh)
DK (1) DK3355919T3 (zh)
EA (1) EA201890850A1 (zh)
ES (1) ES2937167T3 (zh)
FI (1) FI3355919T3 (zh)
HK (1) HK1256067A1 (zh)
HR (1) HRP20221530T1 (zh)
HU (1) HUE060762T2 (zh)
IL (2) IL299478A (zh)
LT (1) LT3355919T (zh)
MD (1) MD3355919T2 (zh)
PL (1) PL3355919T3 (zh)
PT (1) PT3355919T (zh)
SI (1) SI3355919T1 (zh)
WO (1) WO2017055613A2 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210054060A1 (en) * 2012-09-26 2021-02-25 Julius-Maximillians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI710377B (zh) 2017-05-23 2020-11-21 丹麥商諾佛 儂迪克股份有限公司 Mic-1化合物及其用途
WO2021111636A1 (ja) * 2019-12-06 2021-06-10 大塚製薬株式会社 抗gdf15抗体
CN112162097B (zh) * 2020-07-24 2022-04-01 广州医科大学 Gdf1作为评估pd-1单抗治疗效果的生物标志物
IL302646A (en) * 2020-11-10 2023-07-01 Catalym Gmbh Anti-GDF15 antibody and dosage regimen for cancer treatment
CN112778419A (zh) * 2021-02-01 2021-05-11 重庆中元汇吉生物技术有限公司 抗ck-mb的抗体或其抗原结合部分及其应用
CA3215737A1 (en) 2021-03-31 2022-10-06 Cambridge Enterprise Limited Therapeutic inhibitors of gdf15 signalling
WO2023018803A1 (en) 2021-08-10 2023-02-16 Byomass Inc. Anti-gdf15 antibodies, compositions and uses thereof
WO2023122213A1 (en) 2021-12-22 2023-06-29 Byomass Inc. Targeting gdf15-gfral pathway cross-reference to related applications
WO2024052532A1 (en) 2022-09-08 2024-03-14 Catalym Gmbh Anti-gdf15 antibody used in a combination treatment of specific patient groups and a dosage regimen for the treatment of cancer

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005099746A1 (en) 2004-04-13 2005-10-27 St Vincent's Hospital Sydney Limited Method for modulating appetite
US20060148709A1 (en) 1999-05-17 2006-07-06 Biopharm Gesellschaft Zur Biotechnologischen Entwicklung Von Pharmaka Mbh Neuroprotective properties of GDF-15, a novel member of the TGF-Beta superfamily
US20070128636A1 (en) 2005-12-05 2007-06-07 Baker Joffre B Predictors Of Patient Response To Treatment With EGFR Inhibitors
WO2009021293A1 (en) 2007-08-16 2009-02-19 St Vincent's Hospital Sydney Limited Agents and methods for modulating macrophage inhibitory cytokine (mic-1) activity
US8003099B2 (en) 2008-09-29 2011-08-23 Roche Palo Alto Llc Antibodies against human IL17 and uses thereof
WO2011127219A1 (en) 2010-04-06 2011-10-13 Caris Life Sciences Luxembourg Holdings Circulating biomarkers for disease
US20110262444A1 (en) 2010-03-03 2011-10-27 Detroit R&D, Inc. Form-specific antibodies for nag-1 (mic-1, gdf-15), h6d and other tgf-beta subfamily and heart disease and cancer diagnoses
WO2012162561A2 (en) 2011-05-24 2012-11-29 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
WO2013012648A1 (en) 2011-07-15 2013-01-24 Emory University Gdf15 in diagnostic and therapeutic applications
WO2014049087A1 (en) 2012-09-26 2014-04-03 Julius-Maximilians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15)
WO2014100689A1 (en) 2012-12-21 2014-06-26 Aveo Pharmaceuticals, Inc. Anti-gdf15 antibodies
US20140271546A1 (en) 2013-03-15 2014-09-18 Myriad Genetics, Inc. Genes and gene signatures for diagnosis and treatment of melanoma
US20140378665A1 (en) 2012-01-26 2014-12-25 Amgen Inc. Growth differentiation factor 15 (gdf-15) polypeptides
WO2015108907A2 (en) 2014-01-14 2015-07-23 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of melanoma using pd-l1 isoforms
WO2015144855A1 (en) 2014-03-26 2015-10-01 Julius-Maximilians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
WO2016049470A1 (en) 2014-09-25 2016-03-31 Aveo Pharmaceuticals, Inc. Methods of reversing cachexia and prolonging survival comprising administering a gdf15 modulator and an anti-cancer agent

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150104470A1 (en) * 2013-10-11 2015-04-16 Neil H. Riordan Immune modulation by peri-lymphatic or intra-lymphatic cell therapy
CA3109905A1 (en) 2018-08-20 2020-02-27 Pfizer Inc. Anti-gdf15 antibodies, compositions and methods of use

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060148709A1 (en) 1999-05-17 2006-07-06 Biopharm Gesellschaft Zur Biotechnologischen Entwicklung Von Pharmaka Mbh Neuroprotective properties of GDF-15, a novel member of the TGF-Beta superfamily
WO2005099746A1 (en) 2004-04-13 2005-10-27 St Vincent's Hospital Sydney Limited Method for modulating appetite
US20070128636A1 (en) 2005-12-05 2007-06-07 Baker Joffre B Predictors Of Patient Response To Treatment With EGFR Inhibitors
WO2009021293A1 (en) 2007-08-16 2009-02-19 St Vincent's Hospital Sydney Limited Agents and methods for modulating macrophage inhibitory cytokine (mic-1) activity
US20100278843A1 (en) 2007-08-16 2010-11-04 St. Vincent's Hospital Sydney Limited Agents and methods for modulating macrophage inhibitory cytokine (mic-1) activity
US8003099B2 (en) 2008-09-29 2011-08-23 Roche Palo Alto Llc Antibodies against human IL17 and uses thereof
US20110262444A1 (en) 2010-03-03 2011-10-27 Detroit R&D, Inc. Form-specific antibodies for nag-1 (mic-1, gdf-15), h6d and other tgf-beta subfamily and heart disease and cancer diagnoses
WO2011127219A1 (en) 2010-04-06 2011-10-13 Caris Life Sciences Luxembourg Holdings Circulating biomarkers for disease
WO2012162561A2 (en) 2011-05-24 2012-11-29 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
WO2013012648A1 (en) 2011-07-15 2013-01-24 Emory University Gdf15 in diagnostic and therapeutic applications
US20140378665A1 (en) 2012-01-26 2014-12-25 Amgen Inc. Growth differentiation factor 15 (gdf-15) polypeptides
WO2014049087A1 (en) 2012-09-26 2014-04-03 Julius-Maximilians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15)
WO2014100689A1 (en) 2012-12-21 2014-06-26 Aveo Pharmaceuticals, Inc. Anti-gdf15 antibodies
US20140193427A1 (en) 2012-12-21 2014-07-10 Aveo Pharmaceuticals, Inc. Anti-gdf15 antibodies
US20140271546A1 (en) 2013-03-15 2014-09-18 Myriad Genetics, Inc. Genes and gene signatures for diagnosis and treatment of melanoma
WO2015108907A2 (en) 2014-01-14 2015-07-23 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of melanoma using pd-l1 isoforms
WO2015144855A1 (en) 2014-03-26 2015-10-01 Julius-Maximilians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
WO2016049470A1 (en) 2014-09-25 2016-03-31 Aveo Pharmaceuticals, Inc. Methods of reversing cachexia and prolonging survival comprising administering a gdf15 modulator and an anti-cancer agent

Non-Patent Citations (137)

* Cited by examiner, † Cited by third party
Title
Abd El-Aziz et al. "Cleavage of growth differentiation factor 15 (GDF15) by membrane type 1-matrix metalloproteinase abrogates GDF15-mediated suppression of tumor cell growth", Cancer Sci., Sep. 2007, vol. 98, No. 9, pp. 1330-1335.
Adkins, C. et al (2015) "A novel preclinical method to quantitatively evaluate early-stage metastatic events at the murine blood-brain barrier," Cancer Prev Res (Phila) 8(1):68-76.
Altschul et al. (1990) "Basic local alignment search tool," Journal of Molecular Biology 215:403-410.
Altschul et al.: (1997) "Gapped BLAST and PSI-BLAST: a new generation of protein database search programs," Nucleic Acids Res. 25:3389-3402.
Angell, H. et al. (2013) "From the immune contexture to the Immunoscore: the role of prognostic and predictive immune markers in cancer," Curr Opin Immunol. (2):261-7.
Arbabi, G. et al. (1997) "Selection and identification of single domain antibody fragments from camel heavy-chain antibodies." FEBS Lett. 414(3):521-6.
Artz, A. et al. (2016) "GDF-15 inhibits integrin activation and mouse neutrophil recruitment through the ALK-5/TGF-βRII heterodimer," Blood. 128(4):529-41.
Baek et al. "Upregulation and secretion of macrophage inhibitory cytokine-1 (MIC-1) in gastric cancers", Clinica Chimica Acta, 2009, vol. 401, pp. 128-133, doi: 10.1016/j.cca.2008.12.008.
Baek et al., "Nonsteroidal Anti-Inflammatory Drug-Activated Gene-1 Over Expression in Transgenic Mice Suppresses Intestinal Neoplasia", Gastroenterology, 2006, vol. 131, pp. 1553-1560.
Bauskin et al., "The TGF-ß Superfamily Cytokine MIC-1/GDF15: Secretory Mechanisms Facilitate Creation of Latent Stromal Stores", Journal of Interferon & Cytokine Research, 2010, vol. 30, No. 6, pp. 27-35.
Bauskin, A. et al. (2005) "The propeptide mediates formation of stromal stores of PROMIC-1: role in determining prostate cancer outcome," Cancer Res. 65(6):2330-6.
Blanco-Calvo et al., "Circulating levels of GDF15, MMP7 and miR-200c as a poor prognostic signature in gastric cancer", Future Oncology, 2014, vol. 10, No. 7, pp. 1187-1202.
Boehm: Nivolumab beim Nierenzellkarzinom in der Zweitlinie—verlängertes Überleben mit Immuntherapie. Medscape. Sep. 28, 2015.
Bootcov et a., "MIC-1, a novel macrophage inhibitory cytokine. Is a divergent member of the TGF-ß superfamily", Proc. Nat'l. Acad. Sci., Oct. 1997, vol. 94, pp. 11514-11519.
Boyle et al., "Macrophage Inhibitory Cytokine-1 Is Overexpressed in Malignant Melanoma and Is Associated with Tumorigenicity", Aug. 28, 2008, vol. 129, pp. 383-391, doi: 10.1038/jid.2008.270.
Brown et al., "MIC-1 Serum Level and Genotype: Associations with Progress and Prognosis of Colorectal Carcinoma", Clinical Cancer Research, Jul. 2003, vol. 9, pp. 2642-2650.
Brown, D. et al. (2009) "Macrophage inhibitory cytokine 1: a new prognostic marker in prostate cancer," Clin Cancer Res. 15(21):6658-64.
Bruzzese et al., "Local and Systemic Protumorigenic Effects of Cancer-Associated Fibroblast-Derived GDF15", Cancer Research, Apr. 29, 2014, vol. 74, No. 13, pp. 3408-3418, doi: 10.1158/0008-5472.CAN-13-2259.
Chen, S. et al. (2007) "Prostate-derived factor as a paracrine and autocrine factor for the proliferation of androgen receptor-positive human prostate cancer cells," The Prostate. 67(5):557-71.
Cheng, P. et al. (2015) "Data mining the Cancer Genome Atlas in the era of precision cancer medicine," Swiss Med Wkly. 145:w14183.
Chothia, C. et al. (1987) "Canonical structures for the hypervariable regions of immunoglobulins," J Mol Biol. 196(4):901-17.
Chothia, C. et al. (1989) "Conformations of immunoglobulin hypervariable regions," Nature. 342(6252):877-83.
Clackson, T et al. (1991) "Making antibody fragments using phage display libraries." Nature. 352(6336):624-8.
Cong M.et al. (2015) Advertorial: "Novel Bioassay to Assess PD-1/PD-L1 Therapeutic Antibodies in Development for Immunotherapy Bioluminescent Reporter-Based PD-1/PD-L1 Blockade Bioassay." (http://www.genengnews.com/gen-articles/advertorial-novel-bioassay-to-assess-pd-1-pd-l1-therapeutic-antibodies-in-development-for-immun/5511/).
Corre et al., "Bioactivity and Prognostic Significance of Growth Differentiation Factor GDF15 Secreted by Bone Marrow Mesenchymal Stem Cells in Multiple Myeloma", Cancer Research, Feb. 2, 2012, vol. 72, No. 6, pp. 1395-1407, doi: 10.1158/0008-5472.CAN-11-0188.
Corre, J. et al. (2013) "Concise Review: Growth Differentitaion Factor 15 in Pathology: A Clinical Role," Stem Cells Translational Medicine. 2:946-952.
Cully, M. (2015) "Combinations with checkpoint inhibitors at wavefront of cancer immunotherapy," Nat Rev Drug Discov. 14(6):374-5.
Darvin et al. (2018) Experimental & Molecular Medicine 30: 165 pp. 1-11. *
Eisenauer et al. (2009) "New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)," Eur. J. Cancer. 45(2):228-47.
Fisher et al., "MIC-1/GDF15 in Barrett's oesophagus and oesophageal adenocarcinoma", British Journal of Cancer, 2015, vol. 112, pp. 1384-1391, doi: 10.1038/bjc.2015.100.
Gajewski, T. et al. (2013) "Cancer immunotherapy strategies based on overcoming barriers within the tumor microenvironment," Curr Opin Immunol. 25(2):268-76.
Galon et al., "Type, Density, and Location of Immune Cells Within Human Colorectal Tumors Predict Clinical Outcome", Science Mag., Sep. 29, 2006, vol. 313, pp. 1960-1993.
Garber, K. (2015) "Predictive biomarkers for checkpoints, first tests approved," Nat Biotechnol. 33(12):1217-1218.
Gentles, A. et al. (2015) "The prognostic landscape of genes and infiltrating immune cells across human cancers," Nat Med. (8):938-945.
Gouttefangeas, C. et al. (2015) "Flow Cytometry in Cancer Immunotherapy: Applications, Quality Assurance and Future," Cancer Immunology: Translational Medicine from Bench to Bedside (N. Rezaei editor). Springer. Chapter 25: pp. 471-486.
Griner et al., "Growth differentiation factor 15 stimulates rapamycin-sensitive ovarian cancer cell growth and invasion", Biochemical Pharmacology, vol. 85, pp. 46-58.
Guidicelli, V. et al. (2004) IMGT/V-QUEST, an integrated software program for immunoglobulin and T cell receptor V-J and V-D-J rearrangement analysis. Nucleic Acids Res. 32(Web Server issue):W435-40.
Hadrup et al., "Effector CD4 and CD8 T Cells and Their Role in the Tumor Microenvironment", Cancer Microenvironment, 2013, vol. 6, pp. 123-133.
Herbertz, S. et al. (2015) "Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway," Drug Des Devel Ther. 9:4479-99.
Herbst, R. et al. (2014) "Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients," Nature. 515(7528):563-7.
Holliger, P. et al. (1993) "Diabodies: small bivalent and bispecific antibody fragments." Proc Natl Acad Sci U S A. 90(14):6444-8.
Holt, L. et al. (2003) "Domain antibodies: proteins for therapy," Trends Biotechnol. 21(11):484-90.
Huang, C. et al. (2007) "Molecular alterations in prostate carcinomas that associate with in vivo exposure to chemotherapy: identification of a cytoprotective mechanism involving growth differentiation factor 15." Clin Cancer Res.13(19):5825-33.
Huh et al., "Macrophage Inhibitory Cytokine-1 Regulates Melanoma Vascular Development", The American Journal of Pathology, Jun. 2010, vol. 176, No. 6, pp. 2948-2957.
Huh, S. et al. (2010) "Macrophage inhibitory cytokine-1 regulates melanoma vascular development," Am J Pathol. 176 (6):2948-57.
Husaini et al., "Macrophage Inhibitory Cytokine-1 (MIC-1/GDF15) Slows Cancer Development but Increases Metastases in TRAMP Prostate Cancer Prone Mice", PLOS One, Aug. 2012, vol. 7, No. 8, pp. 1-9.
International Search Report and Written Opinion for PCT/EP16/73520.
Jackson, A. et al. (2010) "Recognizing and avoiding siRNA off-target effects for target identification and therapeutic application," Nat Rev Drug Discov. 9(1):57-67.
Jerby-Arnon et al., "A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade", Cell, vol. 175, pp. 984-997, Nov. 1, 2018, doi: 10.1016/j.cell.2018.09.006.
Ji et al., "Twist promotes invasion and cisplatin resistance in pancreatic cancer cells through growth differentiation factor 15", Molecular Medicine Reports, 2015, vol. 12, pp. 3841-3848.
Ji, R. (2011) "An immune-active tumor microenvironment favors clinical response to ipilimumab," Cancer Immunol Immunother. 61(7):1019-31.
Johnen, H et al. (2007) "Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-1," Nat Med. 13(11):1333-40.
Jones et al., "Supraphysiologic Administration of GDF11 Induces Cachexia in Part by Upregulating GDF15", Cell Reports, 2018, vol. 22, pp. 1522-1530.
Jones, P. et al. (1986) "Replacing the complementarity-determining regions in a human antibody with those from a mouse." Nature. 321(6069):522-5.
Joshi et al., "Growth differentiation factor 15 (GDF15)-mediated HER2 phosphorylation reduces trastuzumab sensitivity of HER2-overexpressing breast cancer cells", Biochemical Pharmacology, 2011, vol. 82, pp. 1090-1099.
Joshi, J. et al. (2011) "Growth Differentiation Factor 15 (GDF15)-Mediated HER2 Phosphorylation Reduces Trastuzumab Sensitivity of HER2-Overexpressing Breast Cancer Cells," Biochem Pharmacol.82(9): 1090-1099.
Junker, M. (2015) Development and characterization of monoclonal antibodies to GDF-15 for potential use in cancer therapy. Ph.D. Thesis.
Kanasty, R. et al. (2013) "Delivery materials for siRNA therapeutics," Nat Mater. 12(11):967-77.
Kang et al., "Tolfenamic Acid Induces Apoptosis and Growth Inhibition in Head and Neck Cancer: Involvement of NAG-1 Expression", PLOS One, Apr. 2012, vol. 7, No. 4, pp. 1-10.
Kempf, T. et al. (2011) "GDF-15 is an inhibitor of leukocyte integrin activation required for survival after myocardial infarction in mice," Nat Med.17(5):581-588.
Kim et al., "Implication of NAG-1 in synergistic induction of apoptosis by combined treatment of sodium salicylate and PI3K/MEK1/2 inhibitors in A549 human lung adenocarcinoma cells", Biochemical Pharmacology, 2008, vol. 75, pp. 1751-1760.
Kim et al., "Macrophage inhibitory cytokine-1 activates AKT and ERK-1/2 via the transactivation of ErbB2 in human breast and gastric cancer cells", Carcinogenesis, 2008, vol. 29, No. 4, pp. 704-712.
Kim et al., "NSAID-activated gene 1 mediates pro-inflammatory signaling activation and paclitaxel chemoresistance in type I human epithelial ovarian cancer stem-like cells", Oncotarget, Sep. 30, 2016, vol. 7, No. 44, pp. 72148-72166.
Knoepfel, S. et al. (2012) "Selection of RNAi-based inhibitors for anti-HIV gene therapy," World J Virol. 1(3):79-90.
Köhler, G. (1975) "Continuous cultures of fused cells secreting antibody of predefined specificity," Nature. 256 (5517):495-7.
Lasithiotakis, K. et al. (2006) "The incidience and mortality of cutaneous melanoma in southern Germany," Cancer. 107(6):1331-9.
Lavaud, P. et al. (2014) "Strategies to overcome trastuzumab resistance in HER2-overexpressing breast cancers: focus on new data from clinical trials," BMC Medicine 12:132, 10 pp.
Li et al., "GDF15 promotes EMT and metastasis in colorectal cancer", Oncotarget, Oct. 22, 2015, vol. 7, No. 1, pp. 860-872.
Li et al., "Growth differentiation factor 15 is a promising diagnostic and prognostic biomarker in colorectal cancer", J. Cell. Mol. Med., 2016, vol. 20, No. 8, pp. 1420-1426.
Li, B et al. (2011) "RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome," BMC Bioinformatics. 12:323, 16 pp.
Liu et al., "Association of Serum Level Growth Differentiation Factor 15 with Liver Cirrhosis and Hepatocellular Carcinoma", PLOS One, May 21, 2015, vol. 10, No. 5, pp. 1-13.
Llopiz et al. (2008) "Combined immunization with adjuvant molecules poly(I:C) and anti-CD40 plus a tumor antigen has potent prophylactic and therapeutic antitumor effects," Cancer Immunology & Immunotherapy. 57(1):19-29.
Long et al., "Epacadostat Plus Pembrolizumab Versus Placebo Plus Pembrolizumab in Patients With Unresectable or Metastatic Melanoma (ECHO-301-KEYNOTE-252): A Phase 3, Randomised, Double-Blind Study", Lancet Oncology, vol. 20, No. 8, pp. 1083-1097, Aug. 2019, doi: 10.1016/S1470-2045(19)30274-8.
Maasho et al., "Cutting Edge: NKG2D Is a Costimulatory Receptor for Human Naive CD8+ T Cells", The Journal of Immunology, 2005, vol. 174, No. 8, pp. 4480-4484.
Maier, J. et al. (2015) "Knockdown of platinum-induced growth differentiation factor 15 abrogates p27-mediated tumor growth delay in the chemoresistant ovarian cancer model A2780cis," Cancer Med. 4(2):253-67.
Marks, J. et al. (1991) "By-passing immunization: Human antibodies from V-gene libraries displayed on phage." J Mol Biol. 222(3):581-97.
Mehta et al., "A Prospective Study of Macrophage Inhibitory Cytokine-1 (MIC-1/GDF15) and Risk of Colorectal Cancer", JNCI, Apr. 9, 2014, vol. 106, No. 4, pp. 1-8.
Mehta et al., "Association Between Plasma Levels of Macrophage Inhibitory Cytokine-1 Before Diagnosis of Colorectal Cancer and Mortality", Gastroenterology, 2015, vol. 149, pp. 614-622.
Miller, J. et al. (2015) "The journey from discoveries in fundamental immunology to cancer immunotherapy," Cancer Cell. 27(4):439-49.
Mimeault, K. et al. (2010) "Divergent molecular mechanisms underlying the pleiotropic functions of macrophage inhibitory cytokine-1 in cancer," J Cell Physiol. 224(3):626-35.
Mimeault, M et al. (2013) "Marked improvement of cytotoxic effects induced by docetaxel on highly metastatic and androgen-independent prostate cancer cells by downregulating macrophage inhibitory cytokine-1." Br J Cancer. 108 (5):1079-91.
Motz, G. et al. (2014) "Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors," Nat Med. 20(6):607-15.
Motzer, R. et al. (2015) "Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma," N Engl J Med. 373 (19):1803-13.
Neuzillet, C. et al. (2015) "Targeting the TGFβ pathway for cancer therapy," Pharmacol Ther.147:22-31.
Patel et al., "GDF15 Provides an Endocrine Signal of Nutritional Stress in Mice and Humans", Cell Metabolism, 2019, vol. 29+, pp. 707-718.
Peng et al., "Growth and differentiation factor 15 regulates PD-LI expression in glioblastoma", Cancer Management and Research, 2019, vol. 11, pp. 2653-2661.
Reardon, D. et al. (2013) "An Update of Vaccine Therapy and Other Immunotherapeutic Approaches to Glioblastoma," Expert Rev Vaccines. 12(6):597-615.
Ribas, A. et al. (2015) "Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial," Lancet Oncol. 16:908-18.
Riechmann, L. et al. (1988) "Reshaping human antibodies for therapy," Nature. 332(6162):323-7.
Rizvi, N. et al. (2015) "Cancer immunology: Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer," Science. 348(6230):124-8.
Robert, C. et al. (2015) "Pembrolizumab versus Ipilimumab in Advanced Melanoma," N Engl J Med. 372:2521-2532.
Roth et al., "GDF-15 Contributes to Proliferation and Immune Escape of Malignant Glinomas", Clinical Cancer Research, Jun. 9, 2010, vol. 16, pp. 3851-3860.
Roth, P. et al. (2010) "GDF-15 contributes to proliferation and immune escape of malignant gliomas," Clin Cancer Res. 16(15):3851-9.
Rothschild, S. et al. (2016) "SAKK 16/14: Anti-PD-L1 antibody durvalumab (MEDI4736) in addition to neoadjuvant chemotherapy in patients with stage IIIA(N2) non-small cell lung cancer (NSCLC)—A multicenter single-arm phase II trial," Journal of Thoracic Oncology vol. 11, Suppl. 4S: S106-S112.
Saerens, D et al. (2008) "Single-domain antibodies as building blocks for novel therapeutics," Curr Opin Pharmacol. 8(5):600-8.
Schiegnitz, et al., "GDF 15 as an anti-apoptotic, diagnostic and prognostic marker in oral squamous cell carcinoma", Oral Oncology, 2012, vol. 48, pp. 608-614.
Schiegnitz, et al., "Growth differentiation factor 15 as a radiation-induced marker in oral carcinoma increasing radiation resistance", Journal of Oral Pathology and Medicine, 2016, vol. 45, pp. 63-69.
Search Report Under Section 17 for GB1607801.6.
Sela-Culang et al. (2013) "The structural basis of antibody-antigen recognition," Frontiers in Immunology, vol. 4, Article 302, 13 pp.
Selander et al., "Serum Macrophage Inhibitory Cytokine-1 Concentrations Correlate with the Presence of Prostate Cancer", Cancer Epidemiology, Biomarkers & Prevention, Mar. 2007, vol. 16, No. 3, 532-537.
Senapati et al., "Overexpression of macrophage inhibitory cytokine-1 induces metastasis of human prostate cancer cells through the FAK-RhoA signaling pathway", Oncogene, 2010, vol. 29, pp. 1293-1302.
Senovilla et al., "Prognostic and predictive value of the immune infiltrate in cancer", Trial Watch, Oncolmmunology, 2012, vol. 1, No. 8, pp. 1323-1343.
Shnaper et al., "Elevated levels of MIC-1/GDF15 in the cerebrospinal fluid of patients are associated with glioblastoma and worse outcome", Int. J. Cancer, 2009, vol. 125, pp. 2624-2630.
Siegel, D. (2002) "Recombinant monoclonal antibody technology." Transfus Clin Biol. 9(1):15-22.
Spranger et al., "Tumor-Intrinsic Oncogene Pathways Mediating Immune Avoidance", OncoImmunology, vol. 5, No. 3, e1086862, 2016, doi: 10.1080/2162402X.2015.1086862.
Staff et al., "Elevated Plasma Growth Differentiation Factor-15 Correlates with Lymph Node Metastases and Poor Survival in Endometrial Cancer", Clinical Cancer Research, Jul. 15, 2011, vol. 17, No. 14, pp. 4825-4833.
Staff et al., "Growth differentiation factor-15 as a prognostic biomarker in ovarian cancer", Gynecologic Oncology, 2010, vol. 118, pp. 237-243.
Stefanescu R. et al. (2007) "Mass spectrometric approaches for elucidation of antigenantibody recognition structures in molecular immunology," Eur. J. Mass Spectrom. 13:69-75.
Suchard, S. et al. (2013) "A monovalent anti-human CD28 domain antibody antagonist: preclinical efficacy and safety," J Immunol. 191:4599-4610.
Suckau, D. et al. (1990) "Molecular epitope identification by limited proteolysis of an immobilized antigen-antibody complex and mass spectrometric peptide mapping," Proc Natl Acad Sci U S A. 87(24): 9848-9852.
Tanno et al., "Growth differentiating factor 15 enhances the tumor-initiating and self-renewal potential of multiple myeloma cells", Blood, Jan. 30, 2014, vol. 123, No. 5, pp. 725-733.
Tanno, T. et al. (2010) "Growth differentiation factor 15 in erythroid health and disease," Curr Opin Hematol. 17(3): 184-190.
Tanno, T. et al. (2011) "The TGF-β Family Member Growth Differentiation Factor 15 (GDF15) Regulates the Self-Renewal of Multiple Myeloma Cancer Stem Cells," Blood, 118(21):Abstract 2954.
Taube, J. et al. (2014) "Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy.", Clin Cancer Res. 20(19):5064-74.
Thompson et al., "Human Anti-CD40 Antibody and Poly IC:LC Adjuvant Combination Induces Potent T Cell Responses in the Lung of Nonhuman Primates", The Journal of Immunology, 2015, 195: 1015-1024.
Topalian, S. et al. (2014) "Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab," J. Clin. Oncol. 32(10):1020-1031.
Topalian, S. et al. (2015) "Immune checkpoint blockade: a common denominator approach to cancer therapy," Cancer Cell. 27(4):450-61.
Tsai, K. et al. (2015) "Clinical Chracteristics Predictive of Response to Pembrolizumab uin Advanced Melanoma," J Clin Oncol 33, 2015 (suppl; abstr 9031).
Tsui et al., "Growth differentiation factor-15 upregulates interleukin-6 to promote tumorigenesis of prostate carcinoma PC-3 cells", Journal of Molecular Endocrinology, 2012, vol. 49, pp. 153-163.
Tsui, K. et al. (2012) "Growth differentiation factor-15 upregulates interleukin-6 to promote tumorigenesis of prostate carcinoma PC-3 cells," J Mol Endocrinol. 49(2):153-63.
Tumeh, P. et al. (2014) "PD-1 blockade induces responses by inhibiting adaptive immune resistance," Nature. 515 (7528):568-71.
Van Der Burg, S, et al. (2014) "Immunoguiding, the final frontier in the immunotherapy of cancer." In Cancer Immunotherapy meets oncology (CM Britten, S Kreiter, M. Diken & HG Rammensee eds). Springer International Publishing Switzerland p. 37-51 ISBN: 978-3-319-05103-1.
Wallentin, L. et al. (2013) "GDF-15 for prognostication of cardiovascular and cancer morbidity and mortality in men." PLoS One. 8(12):e78797.
Walsh et al., "NKG2D Receptor Signaling Enhances Cytolytic Activity by Virus-Specific CD8+ T Cells: Evidence for a Protective Role in Virus-Induced Encephalitis", Journal of Virology, Mar. 2008, vol. 82, No. 6, pp. 3031-3044.
Wang et al., "The H6D genetic variation of GDF15 is associated with genesis, progress, and prognosis in colorectal cancer", Pathology—Research and Practice, 2015, vol. 211, pp. 845-850.
Wang, A et al. (2015) "The prognostic value of PD-L1 expression for non-small cell lung cancer patients: a meta-analysis," Eur J Surg Oncol. 41(4):450-6.
Westhrin et al., "Growth differentiation factor 15 (GDF15) promotes osteoclast differentiation and inhibits osteoblast differentiation and high serum GDF15 levels are associated with multiple myeloma bone disease", haematologica, 2015, vol. 100, pp. 511-514.
Xu et al., "Growth differentiation factor 15 induces growth and metastasis of human liver cancer stem-like cells via AKT/GSK-3ß/ß-catenin signaling", Oncotarget, 2017, vol. 8, No. 10, pp. 16972-16987.
Yadav, M. et al. (2014) "Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing," Nature. 515(7528):572-6.
Yang et al., "Elevated level of serum growth differentiation factor 15 is associated with oral leukoplakia and oral squamous cell carcinoma", Journal of Oral Pathology and Medicine, 2014, vol. 43, pp. 28-34.
Yang et al., "GDF 15 is a potential predictive biomarker for TPF induction chemotherapy and promotes tumorigenesis and progression in oral squamous cell carcinoma", Annals of Oncology, 2014, vol. 25, pp. 1215-1222.
Yoon, J. et al. (2013) "Activin receptor-like kinase5 inhibition suppresses mouse melanoma by ubiquitin degradation of Smad4, thereby derepressing eomesodermin in cytotoxic T lymphocytes," EMBO Mol Med.(11):1720-39.
Yoshida et al., "New Immune Checkpoint Inhibitors Human Anti-PD-1 Antibody Nivolumab (Opujibo® intravenous infusion 20 mg, 100 mg) of the Pharmacological Properties and Clinical Efficacy", Japanese Pharmacology Magazine, vol. 146, Issue 2, pp. 106-114, doi: 10.1254/fpj.146.106.
Yu et al., "Tumor-infiltrating T lymphocytes: friends of foes?", Laboratory Investigation, 2006, vol. 86, pp. 231-345.
Zhang, J. et al.(2015) "Prognostic value of pretreatment serum lactate dehydrogenase level in patients with solid tumors: a systematic review and meta-analysis," Scientific Reports 5:9800.
Zhao, L. et al. (2009) "Indication of Candidate Biomarkers of Therapeutic Response to Docetaxel by Proteomic Profiling," Cancer Research. 69(19):7696-7703.
Zhou, Z. et al. (2013) "Growth differentiation factor-15 suppresses maturation and function of dendritic cells and inhibits tumor-specific immune response," PLoS One. 8(11):e78618.

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210054060A1 (en) * 2012-09-26 2021-02-25 Julius-Maximillians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15)
US11891436B2 (en) * 2012-09-26 2024-02-06 Julius-Maximilians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (GDF-15)

Also Published As

Publication number Publication date
WO2017055613A3 (en) 2017-07-06
PL3355919T3 (pl) 2023-03-20
JP6925326B2 (ja) 2021-08-25
DK3355919T3 (da) 2022-12-19
KR20180054868A (ko) 2018-05-24
BR112018006218A2 (pt) 2018-10-09
CA3000293A1 (en) 2017-04-06
PT3355919T (pt) 2023-01-06
FI3355919T3 (fi) 2023-01-13
HRP20221530T1 (hr) 2023-03-17
LT3355919T (lt) 2023-02-10
US20230093412A1 (en) 2023-03-23
AU2023202602A1 (en) 2023-05-18
IL299478A (en) 2023-02-01
ES2937167T3 (es) 2023-03-24
IL258393B2 (en) 2023-08-01
IL258393A (en) 2018-05-31
JP2018534266A (ja) 2018-11-22
JP2021176891A (ja) 2021-11-11
AU2016333539A1 (en) 2018-05-10
HUE060762T2 (hu) 2023-04-28
SI3355919T1 (sl) 2023-04-28
EP4218809A2 (en) 2023-08-02
EA201890850A1 (ru) 2018-09-28
HK1256067A1 (zh) 2019-09-13
JP7443298B2 (ja) 2024-03-05
CN117883572A (zh) 2024-04-16
AU2016333539B2 (en) 2023-02-23
EP3355919A2 (en) 2018-08-08
JP2024054251A (ja) 2024-04-16
CN108463246A (zh) 2018-08-28
EP4218809A3 (en) 2023-08-09
US20190160169A1 (en) 2019-05-30
MD3355919T2 (ro) 2023-02-28
EP3355919B1 (en) 2022-12-07
WO2017055613A2 (en) 2017-04-06
IL258393B1 (en) 2023-04-01

Similar Documents

Publication Publication Date Title
US20230093412A1 (en) Combination therapy using inhibitors of human growth and differentiation factor 15 (gdf-15) and immune checkpoint blockers
US20220242942A1 (en) Gdf-15 as a diagnostic marker to predict the clinical outcome of a treatment with immune checkpoint blockers
US11891436B2 (en) Monoclonal antibodies to growth and differentiation factor 15 (GDF-15)
WISCHHUSEN et al. Patent 3000293 Summary
WISCHHUSEN et al. Sommaire du brevet 3000293
EA044887B1 (ru) Комбинированное лечение с использованием ингибиторов человеческого фактора роста и дифференцировки 15 (gdf-15) и блокаторов контрольных точек иммунного ответа
CN109476759B (zh) Prl3抗体

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

AS Assignment

Owner name: JULIUS-MAXIMILIANS-UNIVERSITAET WUERZBURG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WISCHHUSEN, JOERG;HAAKE, MARKUS;DUMMER, REINHARD;AND OTHERS;SIGNING DATES FROM 20180413 TO 20180427;REEL/FRAME:045843/0261

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY